Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jan 1.
Published in final edited form as: Trends Microbiol. 2012 Oct 11;21(1):6–13. doi: 10.1016/j.tim.2012.09.001

Microbial translocation, immune activation and HIV disease

Nichole R Klatt 1, Nicholas T Funderburg 2, Jason M Brenchley 1
PMCID: PMC3534808  NIHMSID: NIHMS407947  PMID: 23062765

Abstract

The advent of combination antiretroviral therapy (cART) has significantly improved the prognosis of HIV-infected individuals. However, individuals treated long term with cART still manifest increased mortality compared to HIV-uninfected individuals. This increased mortality is closely associated with inflammation, which persists in cART-treated HIV-infected individuals despite levels of plasma viremia being below detection limits. Chronic, pathological immune activation is a key factor in progression to AIDS in untreated HIV-infected individuals. One contributor to immune activation is microbial translocation, which occurs when microbial products traverse the tight epithelial barrier of the gastrointestinal tract. Here we review the mechanisms underlying microbial translocation and its role in contributing to immune activation and disease progression in HIV infection.

Keywords: HIV, Microbial translocation, Immune Activation, Mucosal Immunity

Persistent immune activation and microbial translocation during HIV infection

The pathology of disease caused by human immunodeficiency virus (HIV) infection is complex and multifaceted. In addition to high levels of systemic viral replication, HIV infection results in chronic immune activation and overall immunological dysfunction, which is closely associated with progression to acquired immunodeficiency syndrome (AIDS)1-3. Furthermore, in HIV-infected individuals who are successfully treated with combined antiretroviral therapy (cART), persistent immune activation is associated with increased morbidity and mortality4-6. Several lines of evidence indicate that a key contributor to immune activation and disease progression during HIV infection is microbial translocation6-51. During HIV infection and pathogenic simian immunodeficiency virus (SIV) infection of non-human primates, several factors underlie microbial translocation, including breakdown of the tight epithelial barrier of the gastrointestinal (GI) tract and mucosal immune dysfunction8, 16, 17, 24, 42. A more complete understanding of the mechanisms underlying microbial translocation may provide insight into the development of novel therapeutic interventions aimed at decreasing microbial translocation and subsequent immune activation, thus decreasing morbidity and mortality in HIV infection.

Immunological abnormalities in progressive infection

One of the earliest insults to the immune system after infection with HIV is direct infection and depletion of CD4 T cells52. This depletion first occurs rapidly in mucosal tissues, where high numbers of activated, memory CD4 T cells (the preferred target of HIV) reside. A slower, and progressive, depletion of CD4 T cells in peripheral tissues and blood ensues53. Given that the majority of CD4 T cells reside within mucosal tissues, early loss of mucosal CD4 T cells represents loss of the majority of these cells within the body52. While this depletion likely induces homeostatic pressure to restore mucosal CD4 T cells, low levels of mucosal CD4 T cells are insufficient to cause progression to AIDS53. Indeed, during non-progressive SIV infection of natural hosts, who do not progress to AIDS, moderate depletion of mucosal CD4 T cells is observed, yet does not result in disease progression54, 55.

While CD4 T cells represent the most drastically affected lymphocyte cell type after HIV infection56, other leukocyte subsets are also altered. Indeed, increased turnover, cell cycle perturbations, apoptosis, immune senescence and altered functionality among CD8 T cells, B cells, and innate immune cells are also observed57, 58. Among both CD4 and CD8 T cells, loss of essential naïve and central memory cell populations occurs, resulting in an increased frequency of short-lived effector cells53, 59. Among natural killer (NK) cells, altered functionality, including decreased cytokine production, increased cytotoxicity, and dysfunctional homing has been observed60, 61. Defective B cell responses have also been reported in HIV infection, including hyperactivation of B cells and dysregulation of B cell subsets, leading to altered antibody production62. Dendritic cell (DC) subsets are also significantly altered, including abnormal levels of plasmacytoid and myeloid DCs, and loss of mucosal CD103+ DCs42, 63, 64. Macrophages are also dysfunctional during HIV infection, with a reduced capability for phagocytosing bacterial products in mucosal tissues, and in addition, may be targets for HIV infection16, 29. High plasma levels of proinflammatory cytokines such as interferon-α (IFN-α), interleukin-1 (IL-1), IL-6, IL-18 and tumor necrosis factor-α (TNF-α) are also thought to result from innate cell activation63, 65. Finally, lymphoid tissue fibrosis, manifested by collagen deposition in lymphoid tissues and associated with expression of transforming growth factor-β (TGF-β), limits immunological responses and CD4 T cell restoration, even after initiation of cART66.

These cumulative immunological abnormalities of both the innate and adaptive arms of the immune system have been referred to as immune activation, given that they seem to result from the immune system being overtly activated1. The degree to which the immune system is activated is of extreme importance to HIV-infected individuals considering that it represents the strongest correlate of disease progression, morbidity, and mortality1-5, 67. Thus, chronic, generalized immune activation is far reaching, and it is really no surprise that untreated individuals eventually succumb to opportunistic infections, given the vast dysfunction of the immune system during progressive HIV disease.

The mechanisms which underlie immune activation during HIV infection are multifaceted and complex, and are likely both direct and indirect. One factor which can directly contribute to immune activation is viral replication68. Direct stimulation of the immune system by HIV can occur via at least two mechanisms; stimulation of HIV-specific T and B cells and stimulation of innate cells via viral RNA binding to Toll-like receptor 7 (TLR7) and TLR869. However, the extent to which the immune system is activated during chronic HIV infection cannot solely be attributed to the virus itself. Indeed, the immune compromised state associated with chronic HIV infection can lead to reactivation of viruses normally controlled by the immune system including Epstein Barr virus (EBV) and cytomegalovirus (CMV)68. Replication of these viruses can also contribute to immune activation via recognition of virally infected cells by antigen-specific T and B cells and viral products binding to pattern recognition receptors. One of the direct consequences of activation of the immune system is secretion of proinflammatory cytokines, such as IFN-α, TNF-α, IL-1, IL-6 and IL-18, which can contribute to additional immune activation and apoptosis of immune cells63, 65, 70. High levels of systemic proinflammatory cytokines can also decrease the immune system’s ability to maintain healthy levels of lymphocytes that normally produce homeostatic effector cytokines such as IL-17 and IL-2219, 42. This skewing away from IL-17/IL-22+ lymphocytes likely contributes to immune activation, particularly in mucosal tissues where cells producing effector cytokines such as IL-17 and IL-22 are critical to maintain the integrity of the epithelial barrier19, 42. Indeed, an inability to maintain immunological and epithelial integrity of the GI tract results in translocation of luminal antigens, including microbial products, into peripheral circulation. Microbial products, which can directly stimulate the immune system, in peripheral circulation and other anatomical sites have been shown to contribute to immune activation in HIV-infected individuals and progressively SIV-infected Asian macaques6-10, 12, 14-19, 21, 22, 25, 29, 31, 32, 35, 36, 38, 40, 41, 44, 48-50,71.

Evidence for microbial translocation

The human GI tract is colonized with approximately 1014 normal flora bacteria, which live in symbiosis with the host and can enhance immune function72. These organisms are essential for efficient metabolic function and digestion, and the human body has coevolved with microbiota leading to a symbiotic relationship that promotes a healthy GI tract72. Given the critical importance for these bacteria to avoid direct contact with the systemic immune system, several structural and immunological host factors exist to prevent microbial products from translocating from the lumen of the intestine. These include a physical barrier of epithelial cells16, intestinal IgA73, and a network of immune cells such as macrophages, dendritic cells (DCs), T cells, and innate lymphoid cells42, 72. However, during HIV infection, there is an assault to several of these protective mechanisms, and microbial products translocate into the lamina propria of the GI tract, and, eventually, into systemic circulation.

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 macaques7. Furthermore, the levels of lipopolysachharide (LPS) in these individuals directly correlated with activation of both the adaptive and innate arms of the immune system7. These data provided insights into an underlying cause of immune activation observed during HIV infection. In the six years since these data were published, microbial translocation during pathogenic HIV or SIV infections was corroborated by several groups, with at least 44 reports of microbial translocation occurring during progressive HIV or SIV infection9-55. Microbial translocation is not a phenomena isolated to HIV infection. Indeed, microbial translocation has been reported in several diseases, including inflammatory bowel diseases (IBD), infection by hepatitis C virus (HCV), alcoholism, and cardiovascular disease74.

Microbial translocation and immune activation

While the degree to which microbial translocation, viral replication (e.g. by HIV, CMV, and EBV), and proinflammatory cytokines cause immune activation in progressively HIV or SIV-infected individuals is unclear, several specific effects of microbial product-mediated immune activation can be considered. Indeed, chronic TLR activation in HIV-disease, through recognition of translocated bacterial products and/or viral products, can cause dysregulation of immune responses30, 75 and has been potentially linked to a number of co-morbidities, including dementia in AIDS patients8.

Monocytes and macrophages express a variety of pattern recognition antigen receptors, and stimulation of these cells via these receptors can contribute to inflammation and cardiovascular disease76. Specifically, increased levels of activated monocyte subsets are observed in progressively HIV or SIV-infected individuals and are likely being activated by a variety of microbial products8, 77. Moreover, elevated plasma levels of soluble markers of monocyte and macrophage activation (sCD14 and sCD163) have been reported in HIV infection38, 78. Furthermore, plasma levels of sCD14 (a bacterial LPS receptor) independently predict mortality in cART-treated HIV-infected individuals and plasma levels of sCD163 (a scavenger receptor) are associated with unstable non-calcified coronary plaques in cART-treated HIV-infected individuals38, 78. Thus, chronic innate immune activation may directly contribute to the increased thrombosis risk observed in HIV-infected individuals. Indeed, monocytes and platelets in peripheral blood of HIV-infected individuals express high levels of tissue factor18, 79. Tissue factor can initiate the extrinsic clotting pathway, potentially leading to increased plasma levels of D-dimers (products of fibrinolysis), which have been linked to mortality (all causes) in HIV infected patients4. Moreover, in SIV-infected non-human primates, levels of several coagulation biomarkers, including D-dimers, have been linked to plasma LPS levels; potentially linking thrombosis and cardiovascular disease risk to microbial translocation21, 40.

While recognition of bacterial products by TLR-expressing myeloid cells likely contributes to immune activation and the related co-morbidities observed in HIV-infected individuals, LPS exposure can also contribute to increased T cell turnover in HIV infection. Indeed, increased central memory CD4 T cell turnover is an important aspect of immune activation58 and turnover of CD4 central memory T cell pools may contribute to the depletion of other CD4 T cell populations in progressive HIV and SIV infections59. Interestingly, exposure of peripheral blood mononuclear cells to bacterial TLR ligands in vitro results in increased cell cycling and death of memory CD4 T cells, but not CD8 T cells51, presenting a potential model whereby microbial products may be contributing to increased CD4 T cell turnover in HIV and SIV infection independent of antigen recognition via T cell receptors.

Increased plasma levels of bacterial products are associated with increased turnover of GI tract CD4 T cells and alterations in relative frequencies of individual subsets of GI tract T cells15. Indeed, microbial translocation has been linked to an increased proportion of T regulatory cells compared to T helper 17 (Th17) cells, as well as dysregulation of GI tract γδ T cells in SIV-infected macaques44, 80. Furthermore, low frequencies of GI tract Th17 cells persist in cART-treated, HIV-infected individuals, which is associated with ongoing microbial translocation31. These T cell dysfunctions in the GI tract may be due to decreased homing of T cells to the gut and/or skewing of local T cell populations39, however the precise mechanism(s) that connect T cell subset alterations, T cell turnover and dysfunctionality, damage to the structural barrier of the GI tract and microbial translocation requires further exploration.

SIV infected natural hosts do not progress to AIDS despite high levels of virus replication, and do not have evidence of microbial translocation. This may help explain the lack of immune activation and disease progression in chronically SIV-infected natural hosts7, 55. Furthermore, experimental administration of bacterial products in natural hosts induces immune activation, which is in turn associated with increased viral loads and CD4 T cell depletion81. These data support a role of microbial translocation in immune activation and disease progression during progressive HIV and SIV infections. Indeed, microbial translocation can induce immune activation in the absence of HIV or SIV infections. For example, in pigtail macaques, which have increased levels of damage to their GI tract even in the absence of SIV infection, microbial translocation into the lamina propria of the GI tract correlates with immune activation, both locally in mucosal tissues and systemically21. Furthermore, in idiopathic CD4 lymphocytopenia (ICL), a disorder characterized by low CD4 counts without HIV infection, high CD4 T cell activation directly correlates with increased microbial translocation82. Given these studies, microbial translocation can drive activation of the immune system, even in the absence of chronic virus infection.

Causes of microbial translocation

Several key mechanisms underlying microbial translocation have recently been identified (Figure 1). One defense mechanism which protects against microbial translocation is the structural barrier of the GI tract. The tight epithelial barrier is a single layer of cells which serves to protect the body from translocating bacteria and to absorb luminal nutrients72. During HIV or SIV infection, the integrity of the tight epithelial barrier of the GI tract is compromised, which has been demonstrated by focal breaches in the barrier, decreased expression of epithelial repair tight junction genes, and increased GI tract permeability16, 24, 50, 83. Indeed, focal breaches of the epithelial barrier are in spatial juxtaposition to microbial translocation in vivo, and the extent of damage to the GI tract barrier during HIV and SIV infection is directly associated with the degree of microbial translocation, both locally within the GI tract and systemically16, 24, 50. Similarly, even in the absence of lentiviral infection, damage to the tight epithelial barrier correlates with local levels of LPS, demonstrating the importance of maintaining barrier integrity to avoid microbial translocation21.

Figure 1.

Figure 1

Mechanisms underlying damage to the tight epithelial barrier of the GI tract and microbial translocation during HIV infection. (a) In the absence of HIV infection, retinoic acid produced by CD103+ DCs helps to drive T cells and innate lymphoid cells (ILC) to produce IL-17 and IL-22, which contributes to a healthy epithelial barrier. B cells produce IgA in the lamina propria, which regulate microbial products in the lumen and prohibit microbes (purple rods) from interacting with and translocating across the epithelial barrier. Macrophages (Mφ) are capable of phagocytosing any bacterial products that translocate without a proinflammatory milieu. (b) However, during HIV infection, IL-17 and IL-22 producing lymphocytes are lost, both by direct infection of CD4+ T cells by HIV, and potentially by skewing of homeostatic responses to proinflammatory responses such as IFN-α production by plasmactyoid dendritic cells (pDCs). Further, hyperactivated macrophages no longer phagocytose bacterial products but respond with inflammatory cytokines such as TNF-α and IFN-α. In addition, decreased IgA may lead to decreased control of microbial products. Inflammation and loss of homeostatic cytokines such as IL-17 and IL-22 lead to damage to the tight epithelial barrier of the GI tract, allowing microbial products to translocate and furthering inflammation.

Maintenance of GI tract structural integrity is dependent, in part, on a functional mucosal immune system. Indeed, Th17 cells, which respond to bacterial products and tend to localize to the GI tract, are significantly depleted during HIV and SIV infection, which is associated with GI tract structural damage, microbial translocation and immune activation19, 42, 71. Furthermore, transcriptional profiling of IL-17 producing lymphocytes demonstrates that these cells express several genes important for homeostasis of epithelial cells, including IL-2242. Furthermore, innate lymphoid cells and specialized NK cells that produce IL-17 and IL-22 are associated with a healthy GI tract barrier, and are lost after SIV and HIV infection42, 84. Other mechanisms may contribute to damage to the barrier in addition to loss of IL-17 and IL-22 producing cells in the GI tract. Indeed, HIV replication itself can drive epithelial damage, likely due to the production of proinflammatory cytokines which can directly induce apoptosis of epithelial cells24.

In a healthy individual, if microbes traverse the epithelial barrier and enter the lamina propria, several immune mechanisms exist to kill or transport the bacteria back to the lumen. However, the massive dysfunction of the mucosal immune system during HIV and SIV infection fails to prohibit systemic microbial translocation once microbial products enter the lamina propria (Figure 1). As discussed above, specialized GI tract macrophages are essential in the clearance of microbial products from the lamina propria, however, these cells are dysfunctional during HIV infection18. Another immunological irregularity observed during HIV and SIV infection is B cell dysfunction, including significantly lower levels of IgA62, 85. IgA antibodies are a key component of the mucosal immune system, and in particular are essential in protection against microbial translocation as IgA can directly bind bacteria and prohibit microbes from binding to the epithelium, can neutralize the bioactivity of bacterial products, and IgA can transport bacteria from the lamina propria back into the lumen73.

Taken together, it appears that dysfunction of the GI tract immune system is multifaceted, includes alterations of lymphocyte and myeloid subsets, as well as depletion of CD4 T cells, and this overall dysfunction contributes to microbial translocation. However, whether the immunological abnormalities cause microbial translocation or whether microbial translocation, immune activation, and immunological abnormalities are all dependent upon one another is unclear. Most likely, damage to the epithelial barrier of the GI tract, mucosal immune dysfunction, CD4 T cell depletion, microbial translocation, immune activation, and chronic virus replication all are part of a chronic and perpetual cycle which drive HIV disease progression.

Microbial translocation despite suppressed viremia

Current cART treatments can suppress viremia to undetectable levels for the life of an individual and their use vastly improves the life expectancy of HIV-infected individuals. Further, suppressed viremia diminishes the majority of immune activation contributed to direct HIV replication. However, despite this virus suppression, cART-treated individuals still have increased morbidity and mortality compared to HIV-uninfected individuals, which, is associated with ongoing immune activation4-6. One factor contributing to immune activation despite virus suppression may be ongoing low levels of virus replication from reservoirs that are maintained despite cART. Indeed, complete eradication of HIV is likely impossible even with robust treatments86. Thus, persistence of latently HIV-infected cells may contribute to the ongoing immune activation observed during cART treatment. Another factor which may contribute to immune activation and microbial translocation despite cART is co-infection with other viruses, such as herpes and hepatitis viruses. Indeed, coinfection with hepatitis viruses during HIV infection is associated with higher levels of microbial translocation87.

Microbial translocation likely underlies a substantial amount of the ongoing immune activation in cART-treated individuals. Indeed, while microbial translocation is lower in HIV-infected, cART-treated individuals than untreated individuals, it is significantly higher than in uninfected subjects7. High levels of microbial translocation are associated with immune activation and limited immune reconstitution in HIV-infected pediatric patients on cART88. During cART treatment, individuals can be broadly classified in two groups: immunological responders, which exhibit CD4 T cell reconstitution and slightly decreased immune activation, and immunological non-responders (INR), who maintain high immune activation and low levels of CD4 T cells89-92. Several groups have demonstrated that lack of immune restoration in these INRs is associated with increased levels of microbial translocation10, 12, 26, 36, 45, 93, 94. In addition, the bacterial products which translocate in INRs are polymicrobic and these individuals tend to lose beneficial flora such as the probiotic bacteria Lactobacillaceae36. Furthermore, in cART-treated individuals, there is an inverse correlation between levels of microbial translocation and CD4 T cell reconstitution11, 12, as well as decreased homing of CD4 T cells to the GI tract39. Also, persistence of HIV-infected cells within the GI tract of cART-treated individuals is associated with microbial translocation32. Perhaps the data which most convincingly demonstrate negative effects of microbial translocation are based on findings that levels of microbial translocation independently predict mortality in cART-treated, HIV-infected individuals38.

Ongoing mucosal immune dysfunction during cART treatment likely underlies the microbial translocation observed. Indeed, while cART induces peripheral CD4 T cell reconstitution, this reconstitution is limited in the GI tract52. Collagen deposition in lymphoid tissues of the GI tract is not resolved during cART treatment, which may prohibit GI tract CD4 T cell reconstitution66. However, it may not just be bulk CD4 T cell reconstitution that underlies dysfunctional mucosal immunity, as Th17 cells, which are essential for maintaining homeostasis of epithelial cells, are not reconstituted despite cART, which may exacerbate the ongoing GI tract dysfunctions71. Moreover, during cART, damage to the tight epithelial barrier of the GI tract still occurs, including decreased epithelial repair genes and increased enterocyte apoptosis95. Thus, despite suppression of viral replication by cART, mucosal immune dysfunctions persist and likely underlie microbial translocation.

Therapeutic interventions to decrease microbial translocation

Given the GI tract dysfunctions that lead to microbial translocation and immune activation despite cART, in order to decrease morbidity and mortality in cART-treated individuals, therapeutic interventions aimed at improving GI tract function should be investigated (Box 1). Indeed, several studies have demonstrated promising results in enhancing mucosal immunity during HIV and SIV infection. For example, PD-1 blockade reduces immune activation and microbial translocation by decreasing type-I IFN signaling in the GI tract96. A particularly exciting area of research is modulation of the composition of the GI tract microbiota, and effects of probiotic administration during HIV and SIV infection. Several studies in mice and humans have demonstrated that probiotics enhance mucosal immunity97. Indeed, probiotic administration in HIV-infected individuals has shown promising results with modest improvement in CD4 T cell levels in the periphery, as well as decreased GI disorders98, 99. Given that loss of IL-17 and IL-22 producing lymphocytes is associated with damage to the GI tract42, future studies aimed at increasing these cell populations in mucosal tissues will be essential. Other potential interventions to enhance these cells may be treatment with retinoic acid or probiotic treatments that include increased segmented filamentous bacteria species, both which have been shown to induce IL-17 production by T cells100, 101. Furthermore, enhancing the epithelial barrier itself will be essential, with candidates such as probiotics and glucagon-like peptides, which have shown to enhance epithelial barrier integrity in mice102.

Use of mucosal adjuvants in vaccination strategies may also provide improved GI tract immunity. Indeed, use of Escherichia coli enterotoxin with HIV DNA therapeutic vaccines reduces viral burden in both the GI tract and periphery103. Furthermore, administration of HIV virus like particles with CCL28, which recruits IgA-secreting plasma cells in the GI tract lamina propria, induces enhanced IgA responses in the gut104, which may be essential for improving mucosal immunity. In addition, vaccination using Clostridium perfringens expressing HIV-1 Gag induced robust HIV-specific responses in the GI tract105. Improving mucosal immunity and decreasing damage to the tight epithelial barrier of the GI tract through either treatment strategies or therapeutic vaccination may decrease morbidity and mortality in HIV-infected individuals, thus future studies into potential treatments will be essential.

Concluding remarks

During HIV infection, the strongest predictor of disease progression is immune activation, which persists despite cART and is associated with morbidity and mortality. One cause of immune activation is microbial translocation, which occurs during HIV infection and is associated with driving immune activation and mortality even during cART treatment. Altered mucosal immunity and a damaged tight epithelial barrier of the GI tract likely underlie microbial translocation during HIV infection, and developing therapeutic interventions aimed at restoring GI tract immunity and integrity may decrease HIV disease progression.

Box 1. Outstanding questions.

  • How much immune activation is directly caused by microbial translocation during HIV infection?

  • When does microbial translocation begin during HIV infection?

  • What underlies dissemination of microbial products locally versus systemically?

  • Which host factors are involved in control of microbial translocation both locally and systemically?

  • Does dysbiosis play a role in microbial translocation?

  • What therapeutic interventions may decrease microbial translocation?

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

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 T cells 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.Hunt PW, 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;197:126–133. doi: 10.1086/524143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.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]
  • 5.Rodger AJ, et al. Activation and coagulation biomarkers are independent predictors of the development of opportunistic disease in patients with HIV infection. J Infect Dis. 2009;200:973–983. doi: 10.1086/605447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lichtfuss GF, et al. Biomarkers of immune dysfunction following combination antiretroviral therapy for HIV infection. Biomark Med. 2011;5:171–186. doi: 10.2217/bmm.11.15. [DOI] [PubMed] [Google Scholar]
  • 7.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]
  • 8.Ancuta P, et al. Microbial translocation is associated with increased monocyte activation and dementia in AIDS patients. PLoS ONE. 2008;3:e2516. doi: 10.1371/journal.pone.0002516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Balagopal A, et al. Human immunodeficiency virus-related microbial translocation and progression of hepatitis C. Gastroenterology. 2008 Jul;135:226–233. doi: 10.1053/j.gastro.2008.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.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]
  • 11.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 Dec;46:367–370. doi: 10.1016/j.jcv.2009.09.011. [DOI] [PubMed] [Google Scholar]
  • 12.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]
  • 13.Arias JF, et al. High systemic levels of interleukin-10, interleukin-22 and C-reactive protein in Indian patients are associated with low in vitro replication of HIV-1 subtype C viruses. Retrovirology. 2010;7:15. doi: 10.1186/1742-4690-7-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cassol E, et al. Persistent microbial translocation and immune activation in HIV-1-infected South Africans receiving combination antiretroviral therapy. J Infect Dis. 2010 Sep 1;202:723–733. doi: 10.1086/655229. [DOI] [PubMed] [Google Scholar]
  • 15.Ciccone EJ, et al. Cycling of gut mucosal CD4+ T cells decreases after prolonged anti-retroviral therapy and is associated with plasma LPS levels. Mucosal Immunol. 2010;3:172–181. doi: 10.1038/mi.2009.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.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]
  • 17.Favre D, 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;2:32ra36. doi: 10.1126/scitranslmed.3000632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Funderburg NT, et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood. 2010;115:161–167. doi: 10.1182/blood-2009-03-210179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gordon SN, et al. Disruption of intestinal CD4+ T cell homeostasis is a key marker of systemic CD4+ T cell activation in HIV-infected individuals. J Immunol. 2010;185:5169–5179. doi: 10.4049/jimmunol.1001801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kamat A, et al. Serological markers for inflammatory bowel disease in AIDS patients with evidence of microbial translocation. PLoS One. 2010;5:e15533. doi: 10.1371/journal.pone.0015533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Klatt NR, et al. Compromised gastrointestinal integrity in pigtail macaques is associated with increased microbial translocation, immune activation, and IL-17 production in the absence of SIV infection. Mucosal Immunol. 2010;3:387–398. doi: 10.1038/mi.2010.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Leinert C, et al. Microbial translocation in simian immunodeficiency virus (SIV)-infected rhesus monkeys (Macaca mulatta) J Med Primatol. 2010;39:243–251. doi: 10.1111/j.1600-0684.2010.00429.x. [DOI] [PubMed] [Google Scholar]
  • 23.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]
  • 24.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]
  • 25.Nowroozalizadeh S, et al. Microbial translocation correlates with the severity of both HIV-1 and HIV-2 infections. J Infect Dis. 2010;201:1150–1154. doi: 10.1086/651430. [DOI] [PubMed] [Google Scholar]
  • 26.Rajasuriar R, et al. Biological determinants of immune reconstitution in HIV-infected patients receiving antiretroviral therapy: the role of interleukin 7 and interleukin 7 receptor alpha and microbial translocation. J Infect Dis. 2010;202:1254–1264. doi: 10.1086/656369. [DOI] [PubMed] [Google Scholar]
  • 27.Said EA, et al. Programmed death-1-induced interleukin-10 production by monocytes impairs CD4+ T cell activation during HIV infection. Nat Med. 2010;16:452–459. doi: 10.1038/nm.2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Smith AJ, et al. A role for syndecan-1 and claudin-2 in microbial translocation during HIV-1 infection. J Acquir Immune Defic Syndr. 2010;55:306–315. doi: 10.1097/QAI.0b013e3181ecfeca. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wallet MA, et al. Microbial translocation induces persistent macrophage activation unrelated to HIV-1 levels or T-cell activation following therapy. AIDS. 2010;24:1281–1290. doi: 10.1097/QAD.0b013e328339e228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bukh AR, et al. Endotoxemia is associated with altered innate and adaptive immune responses in untreated HIV-1 infected individuals. PLoS One. 2011;6:e21275. doi: 10.1371/journal.pone.0021275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.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]
  • 32.d’Ettorre G, et al. HIV persistence in the gut mucosa of HIV-infected subjects undergoing antiretroviral therapy correlates with immune activation and increased levels of LPS. Curr HIV Res. 2011 Apr;9:148–153. doi: 10.2174/157016211795945296. [DOI] [PubMed] [Google Scholar]
  • 33.Kramski M, et al. Novel sensitive real-time PCR for quantification of bacterial 16S rRNA genes in plasma of HIV-infected patients as a marker for microbial translocation. J Clin Microbiol. 2011;49:3691–3693. doi: 10.1128/JCM.01018-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lim A, et al. Antibody and B-cell responses may control circulating lipopolysaccharide in patients with HIV infection. AIDS. 2011;25:1379–1383. doi: 10.1097/QAD.0b013e328348a789. [DOI] [PubMed] [Google Scholar]
  • 35.Marchetti G, et al. Microbial translocation predicts disease progression of HIV-infected antiretroviral-naive patients with high CD4+ cell count. AIDS. 2011 Jul 17;25:1385–1394. doi: 10.1097/QAD.0b013e3283471d10. [DOI] [PubMed] [Google Scholar]
  • 36.Merlini E, et al. Evidence for polymicrobic flora translocating in peripheral blood of HIV-infected patients with poor immune response to antiretroviral therapy. PLoS One. 2011;6:e18580. doi: 10.1371/journal.pone.0018580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Papasavvas E, et al. Increased microbial translocation in </=180 days old perinatally human immunodeficiency virus-positive infants as compared with human immunodeficiency virus-exposed uninfected infants of similar age. Pediatr Infect Dis J. 2011 Oct;30:877–882. doi: 10.1097/INF.0b013e31821d141e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.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]
  • 39.Mavigner M, et al. Altered CD4+ T cell homing to the gut impairs mucosal immune reconstitution in treated HIV-infected individuals. J Clin Invest. 2012;122:62–69. doi: 10.1172/JCI59011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Pandrea I, et al. Coagulation biomarkers predict disease progression in SIV-infected nonhuman primates. Blood. 2012 Aug 16;120:1357–1366. doi: 10.1182/blood-2012-03-414706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pilakka-Kanthikeel S, et al. Increased gut microbial translocation in HIV-infected children persists in virologic responders and virologic failures after antiretroviral therapy. Pediatr Infect Dis J. 2012;31:583–591. doi: 10.1097/INF.0b013e31824da0f5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.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 doi: 10.1038/mi.2012.38. doi: 10.1038/mi.2012.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Papasavvas E, et al. Delayed loss of control of plasma lipopolysaccharide levels after therapy interruption in chronically HIV-1-infected patients. AIDS. 2009;23:369–375. doi: 10.1097/QAD.0b013e32831e9c76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Harris LD, et al. Mechanisms underlying {gamma}{delta} T cell subset perturbations in SIV-infected Asian rhesus macaques. Blood. 2010 Nov 18;116:4148–4157. doi: 10.1182/blood-2010-05-283549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Troseid M, et al. Elevated plasma levels of lipopolysaccharide and high mobility group box-1 protein are associated with high viral load in HIV-1 infection: reduction by 2-year antiretroviral therapy. AIDS. 2010;24:1733–1737. doi: 10.1097/QAD.0b013e32833b254d. [DOI] [PubMed] [Google Scholar]
  • 46.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 Aug 15;57:363–370. doi: 10.1097/QAI.0b013e31821a603c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lyons JL, et al. Plasma sCD14 is a biomarker associated with impaired neurocognitive test performance in attention and learning domains in HIV infection. J Acquir Immune Defic Syndr. 2011;57:371–379. doi: 10.1097/QAI.0b013e3182237e54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chaoul N, et al. Default in plasma and intestinal IgA responses during acute infection by Simian Immunodeficiency Virus. Retrovirology. 2012;9:43. doi: 10.1186/1742-4690-9-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dyavar Shetty R, et al. PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques. J Clin Invest. 2012;122:1712–1716. doi: 10.1172/JCI60612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mohan M, et al. Focused examination of the intestinal lamina propria yields greater molecular insight into mechanisms underlying SIV induced immune dysfunction. PLoS One. 2012;7:e34561. doi: 10.1371/journal.pone.0034561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Funderburg N, et al. Toll-like receptor ligands induce human T cell activation and death, a model for HIV pathogenesis. PLoS One. 2008;3:e1915. doi: 10.1371/journal.pone.0001915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Brenchley JM, Douek DC. HIV infection and the gastrointestinal immune system. Mucosal Immunol. 2008;1:23–30. doi: 10.1038/mi.2007.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Douek DC, et al. T cell dynamics in HIV-1 infection. Annu Rev Immunol. 2003;21:265–304. doi: 10.1146/annurev.immunol.21.120601.141053. [DOI] [PubMed] [Google Scholar]
  • 54.Gordon SN, et al. Severe depletion of mucosal CD4+ T cells in AIDS-free simian immunodeficiency virus-infected sooty mangabeys. J Immunol. 2007;179:3026–3034. doi: 10.4049/jimmunol.179.5.3026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Paiardini M, et al. Lessons learned from the natural hosts of HIV-related viruses. Annual Review of Medicine. 2009;60:485–495. doi: 10.1146/annurev.med.60.041807.123753. [DOI] [PubMed] [Google Scholar]
  • 56.Klatt NR, Silvestri G. CD4+ T Cells and HIV: a paradoxical pas de deux. Science Translational Medicine. 2012;4:123ps124. doi: 10.1126/scitranslmed.3003862. [DOI] [PubMed] [Google Scholar]
  • 57.Ribeiro RM, et al. In vivo dynamics of T cell activation, proliferation, and death in HIV-1 infection: why are CD4+ but not CD8+ T cells depleted? Proc Natl Acad Sci U S A. 2002;99:15572–15577. doi: 10.1073/pnas.242358099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Paiardini M, et al. Cell-cycle dysregulation in the immunopathogenesis of AIDS. Immunol Res. 2004;29:253–268. doi: 10.1385/IR:29:1-3:253. [DOI] [PubMed] [Google Scholar]
  • 59.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]
  • 60.Reeves RK, et al. CD16-natural killer cells: enrichment in mucosal and secondary lymphoid tissues and altered function during chronic SIV infection. Blood. 2010;115:4439–4446. doi: 10.1182/blood-2010-01-265595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Alter G, et al. Increased natural killer cell activity in viremic HIV-1 infection. J Immunol. 2004;173:5305–5311. doi: 10.4049/jimmunol.173.8.5305. [DOI] [PubMed] [Google Scholar]
  • 62.Moir S, Fauci AS. B cells in HIV infection and disease. Nat Rev Immunol. 2009;9:235–245. doi: 10.1038/nri2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kwa S, et al. Plasmacytoid dendritic cells are recruited to the colorectum and contribute to immune activation during pathogenic SIV infection in rhesus macaques. Blood. 2011;118:2763–2773. doi: 10.1182/blood-2011-02-339515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Reeves RK, et al. SIV infection induces accumulation of plasmacytoid dendritic cells in the gut mucosa. J Infect Dis. 2012 doi: 10.1093/infdis/jis408. doi: 10.1093/infdis/jis408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Connolly NC, et al. Proinflammatory cytokines in HIV disease-a review and rationale for new therapeutic approaches. AIDS Rev. 2005;7:168–180. [PubMed] [Google Scholar]
  • 66.Estes JD, et al. The role of collagen deposition in depleting CD4+ T cells and limiting reconstitution in HIV-1 and SIV infections through damage to the secondary lymphoid organ niche. Semin Immunol. 2008;20:181–186. doi: 10.1016/j.smim.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Sodora DL, Silvestri G. Immune activation and AIDS pathogenesis. AIDS. 2008;22:439–446. doi: 10.1097/QAD.0b013e3282f2dbe7. [DOI] [PubMed] [Google Scholar]
  • 68.Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol. 2008;214:231–241. doi: 10.1002/path.2276. [DOI] [PubMed] [Google Scholar]
  • 69.Alter G, et al. Single-stranded RNA derived from HIV-1 serves as a potent activator of NK cells. J Immunol. 2007;178:7658–7666. doi: 10.4049/jimmunol.178.12.7658. [DOI] [PubMed] [Google Scholar]
  • 70.Iannello A, et al. Role of interleukin-18 in the development and pathogenesis of AIDS. AIDS Rev. 2009;11:115–125. [PubMed] [Google Scholar]
  • 71.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]
  • 72.Hooper LV, et al. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273. doi: 10.1126/science.1223490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Macpherson AJ, et al. Homeland security: IgA immunity at the frontiers of the body. Trends Immunol. 2012;33:160–167. doi: 10.1016/j.it.2012.02.002. [DOI] [PubMed] [Google Scholar]
  • 74.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]
  • 75.Lester RT, et al. HIV-1 RNA dysregulates the natural TLR response to subclinical endotoxemia in Kenyan female sex-workers. PLoS One. 2009;4:e5644. doi: 10.1371/journal.pone.0005644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007;81:584–592. doi: 10.1189/jlb.0806510. [DOI] [PubMed] [Google Scholar]
  • 77.Kim WK, et al. Monocyte heterogeneity underlying phenotypic changes in monocytes according to SIV disease stage. J Leukoc Biol. 2009;87:557–567. doi: 10.1189/jlb.0209082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.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]
  • 79.Mayne E, et al. Increased platelet and microparticle activation in HIV infection: upregulation of P-selectin and tissue factor expression. J Acquir Immune Defic Syndr. 2012;59:340–346. doi: 10.1097/QAI.0b013e3182439355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Favre D, et al. Critical loss of the balance between Th17 and T regulatory cell populations in pathogenic SIV infection. PLoS Pathog. 2009;5:e1000295. doi: 10.1371/journal.ppat.1000295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pandrea I, et al. Cutting edge: experimentally induced immune activation in natural hosts of simian immunodeficiency virus induces significant increases in viral replication and CD4+ T cell depletion. J Immunol. 2008;181:6687–6691. doi: 10.4049/jimmunol.181.10.6687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lee PI, et al. Evidence for translocation of microbial products in patients with idiopathic CD4 lymphocytopenia. J Infect Dis. 2009;199:1664–1670. doi: 10.1086/598953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.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]
  • 84.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 Sep 22;118:3321–3330. doi: 10.1182/blood-2011-04-347260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Klatt NR, et al. SIV infection of rhesus macaques results in dysfunctional T- and B-cell responses to neo and recall Leishmania major vaccination. Blood. 2011;118:5803–5812. doi: 10.1182/blood-2011-07-365874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pierson T, et al. Reservoirs for HIV-1: mechanisms for viral persistence in the presence of antiviral immune responses and antiretroviral therapy. Annu Rev Immunol. 2000;18:665–708. doi: 10.1146/annurev.immunol.18.1.665. [DOI] [PubMed] [Google Scholar]
  • 87.Page EE, et al. HIV and hepatitis C coinfection: pathogenesis and microbial translocation. Curr Opin HIV AIDS. 2011;6:472–477. doi: 10.1097/COH.0b013e32834bbc71. [DOI] [PubMed] [Google Scholar]
  • 88.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]
  • 89.Pitrak DL, et al. Discordant CD4 T lymphocyte responses to antiretroviral therapy for HIV infection are associated with ex-vivo rates of apoptosis. AIDS. 2001;15:1317–1319. doi: 10.1097/00002030-200107060-00018. [DOI] [PubMed] [Google Scholar]
  • 90.Hunt PW, et al. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187:1534–1543. doi: 10.1086/374786. [DOI] [PubMed] [Google Scholar]
  • 91.Valdez H, et al. Limited immune restoration after 3 years’ suppression of HIV-1 replication in patients with moderately advanced disease. AIDS. 2002;16:1859–1866. doi: 10.1097/00002030-200209270-00002. [DOI] [PubMed] [Google Scholar]
  • 92.Marchetti G, et al. Comparative analysis of T-cell turnover and homeostatic parameters in HIV-infected patients with discordant immunevirological responses to HAART. AIDS. 2006;20:1727–1736. doi: 10.1097/01.aids.0000242819.72839.db. [DOI] [PubMed] [Google Scholar]
  • 93.Tincati C, et al. Early initiation of highly active antiretroviral therapy fails to reverse immunovirological abnormalities in gut-associated lymphoid tissue induced by acute HIV infection. Antivir Ther. 2009;14:321–330. [PubMed] [Google Scholar]
  • 94.Piconi S, et al. Immune activation, apoptosis, and Treg activity are associated with persistently reduced CD4+ T-cell counts during antiretroviral therapy. AIDS. 2010;24:1991–2000. doi: 10.1097/QAD.0b013e32833c93ce. [DOI] [PubMed] [Google Scholar]
  • 95.Li Q, et al. Functional genomic analysis of the response of HIV-1-infected lymphatic tissue to antiretroviral therapy. J Infect Dis. 2004;189:572–582. doi: 10.1086/381396. [DOI] [PubMed] [Google Scholar]
  • 96.Dyavar Shetty RD, et al. PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques. J Clin Invest. 2012;122:1712–1716. doi: 10.1172/JCI60612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Madsen KL. The use of probiotics in gastrointestinal disease. Can J Gastroenterol. 2001;15:817–822. doi: 10.1155/2001/690741. [DOI] [PubMed] [Google Scholar]
  • 98.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]
  • 99.Hummelen R, et al. 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]
  • 100.Hall JA, et al. Essential role for retinoic acid in the promotion of CD4+ T cell effector responses via retinoic acid receptor alpha. Immunity. 2011;34:435–447. doi: 10.1016/j.immuni.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ivanov II, Littman DR. Segmented filamentous bacteria take the stage. Mucosal Immunol. 2010;3:209–212. doi: 10.1038/mi.2010.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Cani PD, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58:1091–1103. doi: 10.1136/gut.2008.165886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Fuller DH, et al. Therapeutic DNA vaccine induces broad T cell responses in the gut and sustained protection from viral rebound and AIDS in SIV-infected rhesus macaques. PLoS One. 2012;7:e33715. doi: 10.1371/journal.pone.0033715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rainone V, et al. CCL28 induces mucosal homing of HIV-1-specific IgA-secreting plasma cells in mice immunized with HIV-1 virus-like particles. PLoS One. 2011;6:e26979. doi: 10.1371/journal.pone.0026979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Pegu P, et al. Induction of strong anti-HIV cellular immunity by a combination of Clostridium perfringens expressing HIV gag and virus like particles. Curr HIV Res. 2011;9:613–622. doi: 10.2174/157016211798998808. [DOI] [PubMed] [Google Scholar]

RESOURCES