Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Aug 1.
Published in final edited form as: Curr Opin Immunol. 2014 Jul 2;0:127–136. doi: 10.1016/j.coi.2014.06.007

Aging of the Human Innate Immune System in HIV Infection

Heidi J Zapata 1, Albert C Shaw 1
PMCID: PMC4145838  NIHMSID: NIHMS611050  PMID: 24997358

Abstract

HIV infection is associated with a chronic inflammatory state arising from multiple factors, including innate immune recognition of HIV, increased microbial translocation, and release of endogenous ligands from damaged cells (such as CD4 T cells). In many respects, this heightened pro-inflammatory environment resembles that associated with aging in the absence of HIV infection, and evidence of dysregulated innate immune responses can be found in not only older HIV-negative adults, but also adults with HIV infection. While the study of innate immune aging in HIV infection is still in its early stages, it seems likely that at least additive, or potentially synergistic effects of aging and HIV infection will be found.


The life expectancy of a 20-year-old HIV-positive adult on antiretroviral therapy (ART) is expected to be approximately 70 years [1,2], and it is estimated that by 2015, half of HIV-infected individuals in the United States will be over 50 [3]; aging of the HIV-infected population will occur in the developing world with increased availability of ART, and will profoundly affect HIV care. Consequently, understanding the nexus of immune activation from chronic viral infection with age-associated alterations in immune system responsiveness takes on particular urgency [4,5]. Here, we focus on the human innate immune system and the contribution of dysregulated inflammation to the clinical course of older adults with HIV disease.

HIV-Associated Innate Immune Activation: Inflamm-Aging Redux?

The association of HIV infection with a heightened pro-inflammatory environment was described early in the pandemic, with increased plasma levels of neopterin (a byproduct of GTP catabolism associated with macrophage activation), IL-6, TNF-α , TGF-β and others [6]. Elevations in IL-6, D-dimer (a fibrin degradation product that reflects activation of the coagulation cascade), and soluble CD14 (a marker of monocyte activation) were associated with CD4 counts <200 in a cohort of HIV-infected US veterans [7], and several studies have shown that HIV infection is associated with immune activation and elevated coagulation factors [8,9]. Enhanced inflammation is also apparent in elite controllers, a subset of HIV-infected patients who maintain low viral loads in the absence of ART, suggesting that high levels of HIV replication are not required for this heightened pro-inflammatory state [10]. Similar increases in neopterin [11], cytokines, coagulation factors, and acute phase reactants [12] have been reported in older, compared to younger, HIV-negative adults; the term “inflamm-ageing” [13,14] was coined to describe this age-associated pro-inflammatory environment. The intersection of HIV disease and immunosenescence is likely to result in potentiation of age-associated immune dysregulation.

Several studies evaluated effects of age on innate immune activation in HIV-negative versus HIV-positive adults (with the caveat that very few HIV-infected older adults were actually studied). For example, CD14+ CD16+ inflammatory monocytes appear increased in HIV-positive, compared to HIV-negative young adults [15], a finding that has also been reported for older versus young HIV-negative adults [1618]. Monocytes from older adults have increased expression of CD11b (a marker of monocyte activation), and decreased expression of CD38, CD62L, and CD115. Plasma levels of soluble CD163 (sCD163) and CXCL10, both markers of innate immune activation, also increase with age [16]. Notably, these findings were also found in HIV-infected young men [15]; in a cohort of women age 20–63 yrs levels of plasma sCD163 levels in HIV-infected subjects matched levels seen in uninfected women 14.5 years older [19].

Monocyte TLR Function in Aging

TLR function also shows dysregulation in monocytes from older adults and HIV-infected individuals. In older adults, an age-related decrease in TLR1/TLR2-mediated cytokine production was associated with decreased TLR-1 surface expression [18,20]; in addition, generalized age-associated defects in TLR-induced expression of CD80 and CD86 are associated with influenza vaccine response [21]. At the same time, TLR4-induced cytokine production in CD14+ CD16+ inflammatory monocytes was increased with age [16], and TLR5-induced IL-6 and IL-8 production were increased in monocytes isolated by adherence to plastic from older, compared to young adults [22]. Taken together, these findings suggest that activation state could influence age-associated monocyte TLR responses.

Monocyte TLR Function in HIV Infection

Monocyte TLR function in the context of HIV infection generally show evidence of enhanced responsiveness. For example, using cryopreserved PBMC samples, TLR7/8- and TLR2-dependent intracellular TNF-α production were increased in monocytes from subjects with primary HIV infection, compared to HIV+ subjects on ART, elite controllers, or HIV-negative individuals [23]. Analysis of freshly isolated monocytes from chronically infected subjects showed increased expression of TLR2, which was upregulated upon exposure to gp120 [24]. It should be noted that these studies enrolled HIV-infected subjects of a range of ages, but the specific effects of age remain incompletely understood.

Dendritic Cell TLR Function in Aging

Primary dendritic cell function in aging generally shows decreased TLR-dependent cytokine production in both myeloid (mDC) [25,26] and plasmacytoid (pDC) populations (and decreased IFNα production in pDCs) [2630]. Notably, basal cytokine production was markedly elevated in mDCs and pDCs from older, but not young subjects, reflecting the heightened pro-inflammatory environment of aging and suggesting that impaired TLR responses to newly encountered agonists reflect inability to further upregulate cytokine production [26]. However, monocyte-derived DCs, obtained via IL-4 and GM-CSF stimulation, show both increased cytokine production in response to TLR4 and TLR8 engagement and treatment with self-DNA [31,32] and decreased type I interferon production in response to West Nile virus [33]. Thus, as with monocytes, activation state may also play a role in age-associated alterations in TLR function.

Dendritic Cell TLR Function in HIV Infection

TLR function in DCs from HIV-infected subjects may be influenced by stage of infection. For example, TLR8-induced TNF-α production was diminished in mDCs from cryopreserved PBMC samples at the earliest stages of primary infection [34]. However, in later stages of primary disease (when HIV-specific antibodies become detectable—Fiebig stage V–VI), increased TLR-induced cytokine production is found [23,35], although there is also evidence of impaired TLR-induced IL-12 production [36]. By contrast, in chronic HIV infection, decreased TLR-dependent IL-12 production is reported in monocyte-derived DCs [3739]. The mechanisms underlying these alterations are likely to be multifactorial, but HIV-1-induced inhibition of autophagy [40] and release of apoptotic microparticles from dying cells during primary infection may contribute [34].

Diminished numbers of pDC have been reported in the peripheral blood of HIV patients [41,42] and in non-infected older adults [27,43]. Studies of TLR-dependent pDC function in primary and established HIV infection have generally demonstrated decreases in both IFN-α production and induction of NK cell cytotoxicity [4446]. The HIV-1 envelope protein gp120 inhibits TLR9-mediated IFN-α production in pDCs, and so may facilitate this inhibition [47].

Despite lower numbers of pDC, higher plasma levels of IFN-α are found in acute and late stages of HIV infection that are associated with extent of disease progression [48]. Basal transcription of interferon-stimulated genes is increased in pDCs from HIV-and HCV-infected subjects, which could contribute to a heightened baseline level of IFN-α production [45]. In addition, pDCs from HIV-1-infected subjects show persistent IFN-α production in response to repeated in vitro stimulation with HIV-1, associated with increased mRNA expression of IRF7 and incomplete activation as manifested by costimulatory protein expression levels [49]. This HIV-induced dysregulation mirrors the basal elevation in IFN-α production in pDCs from HIV-negative older adults [26].

Sources of Innate Immune Activation in HIV-infected and Uninfected Older Adults

Engagement of Innate Immune Pattern Recognition Receptors by HIV

HIV single-stranded RNA activates TLR7 and TLR8 [5052] on antigen presenting cells; persistent active viral replication (or at least the production of viral RNA) may occur despite treatment with ART, albeit at very low levels [53,54]. In vitro pre-treatment of PBMCs with TLR7 or TLR8 agonists was associated with augmented LPS-induced cytokine production [55,56], suggesting that HIV-dependent TLR responses could contribute to further inflammatory dysregulation. TLR-mediated recognition of HIV may also influence infection acquisition; using TLR neutralizing antibodies, the HIV envelope protein gp120 was found to be recognized by TLR 2 and 4 on cultured human primary genital epithelial cells, and associated with mucosal barrier disruption [57]. How age-associated alterations in TLR function influence HIV-associated TLR activation remains to be determined.

Microbial Translocation in HIV Infection

CCR5+ CD4 T cells in lymphoid tissues in the gastrointestinal tract are depleted in HIV disease, and intestinal mucosa compromise is also observed [58,59]. These factors likely contribute to translocation of gastrointestinal bacteria into the bloodstream, which is associated with innate immune activation and elevated levels of plasma LPS, soluble CD14, LPS binding protein, and IFN-α [60]. Numerous studies indicate that these parameters remain persistently elevated in HIV patients despite ART and correlate with severity of disease [59,61]. The involvement of IFN-α, typically associated with antiviral responses, was highlighted by a recent study indicating that pDCs—which are particularly adept at producing IFN-α—upregulate the gut homing receptor CD103, and accumulate in gut-associated lymphoid tissue in ART-naïve subjects [41]. Additional studies showed higher plasma levels of bacterial DNA in HIV-infected subjects that correlated with plasma LPS levels [62,63]. A few studies have shown differences in the intestinal microbiome of HIV-infected versus uninfected individuals that are associated with immune activation [64,65]. Notably, a study of older HIV-negative adults (mean age of 69) showed higher levels of LPS binding protein (a surrogate marker of microbial translocation) correlated with worsened physical function, raising the intriguing possibility that microbial translocation could also be a feature of advancing age [66].

CMV and Chronic Viral Infections

Following primary infection, the immunologic impact of CMV arises from cycles of asymptomatic latent virus reactivation that may accompany episodes of acute non-viral illness [67]. Increased frequency and heightened reactivation of CMV are also noted in older adults [68]. While CMV seropositive status is associated with innate immune activation [69], it should be noted that there is an absence of longitudinal data supporting CMV-associated innate immune activation. In fact, in a longitudinal study of 249 subjects, individuals who seroconverted to CMV during the 10-year study period did not show a difference in serum CRP, TNF-α, IL-6, and IL-10 compared to those remaining seropositive [70]. It is possible that multiple cycles of reactivation may be required to induce a state of immune activation; additional longitudinal data will be needed to further address this question. In HIV infection, CMV is an important cause of opportunistic infection; in addition, there is evidence implicating a detectable CMV viral load as a risk factor for HIV disease progression independent of CD4 count or viral load [71]. Other chronic, persistent viruses including herpesviruses (e.g. Epstein-Barr, Varicella-Zoster) and Hepatitis C virus likely also contribute to a pro-inflammatory state in HIV infection, but the influence of aging on this interface remains incompletely understood.

Damage-Associated Molecular Patterns

Endogenous DNA damage may also contribute to a pro-inflammatory milieu in the context of aging and HIV infection. In this context, the Senescence-Associated Secretory Phenotype (SASP), representing a secretome of pro-inflammatory cytokines (IL-1β, IL-6, IL-8), growth factors, extracellular matrix proteins, and proteases, is induced by DNA damage from genotoxic stress (e.g. from ionizing radiation or replicative exhaustion) [72,73], though it is worth noting that there is a paucity of data demonstrating an in vivo role for the SASP. However, increased levels of endogenous non-cell-associated DNA were present in a cohort of nonagenarians compared to younger subjects, and correlated with higher expression of inflammatory pathways, frailty, and mortality [74,75]; in addition, increased levels of mitochondrial DNA were associated with monocyte activation and elevated plasma cytokines [76]. This age-associated increase in non-cell-associated DNA could be engaged by innate immune pattern recognition receptors or reflect DNA damage associated with the pro-inflammatory SASP. Endogenous extracellular DNA is likely released from necrotic or senescent cells, and it is notable that necrotic cells activate the NLRP3 inflammasome [7779]. Recent findings propose that bystander CD4+ T cells (which are non-productively infected with HIV) die via the related process of pyroptosis due to the presence of cytosolic reverse transcription products, which is dependent on caspase-1 (a crucial component of the inflammasome mediating the processing of pro-IL-1β and IL-18) [80]. Thus, it seems likely that age-associated increases in cell-free DNA, combined with HIV-1-dependent pyroptosis will augment inflammatory responses in older adults with HIV disease.

Hormonal and Metabolic factors

Estrogen and testosterone treatment result in decreased IL-6 production, and decreases in sex hormone production in menopause or andropause are associated with elevated pro-inflammatory cytokine levels in HIV-negative adults [8186]. Notably, hypogonadism and low testosterone are common among HIV-infected men [87]. Thus, the pro-inflammatory environment in older HIV-positive adults will also be influenced by age-associated changes in sex hormones, and by hormone replacement therapies in older adults.

Adipose tissue represents another potential source for innate immune dysregulation in the context of aging. In the absence of HIV infection, age-associated loss of subcutaneous adipose tissue and increases in intra-abdominal visceral fat coincide with increases in insulin resistance and metabolic syndrome [88]. Adipose tissue is also a source of pro-inflammatory cytokines; increased pro-inflammatory cytokine production has been observed in aged murine adipocytes [8991]. In humans, elevated systemic pro-inflammatory cytokines have been correlated with increased levels of visceral adipose tissue in older adults [9294].

Abnormalities in adipose tissue, or lipodystrophy, associated with HIV have features resembling age-associated fat redistribution, and may consist of a combination of lipoatrophy in the face, extremities and buttocks with hypertrophy of visceral fat depots and cervical fat; lipodystrophy is also associated with increased pro-inflammatory cytokine production in adipocytes [95]. Adverse effects of ART are significant contributors to lipodystrophy via inhibition of mitochondrial DNA polymerase γ (Pol γ), leading to loss of mitochondrial DNA (mtDNA) and increased mtDNA mutations [9699]. Pol γ toxicity was most strongly associated with nucleotide reverse transcriptase inhibitors (NRTIs) no longer in wide use in the developed world, particularly azidothymidine (AZT), stavudine (D4T), and didanosine (DDI) [95,100]. The resultant mtDNA mutations are maintained and may undergo clonal expansion—as reported in muscle biopsies from NRTI-treated compared to treatment naïve subjects [101]. Thus, ART-associated mtDNA mutations may resemble those accumulating in the context of aging [102], and exposure to NRTIs could have long-term consequences beyond the period of actual treatment.

mtDNA content in adipose tissue was also decreased, relative to pre-treatment levels, with ART including widely used agents such as Abacavir/lamivudine (ABC/3TC) or Tenofovir/Emtricitabine (TDF/FTC); TDF/FTC was also associated with decreased enzymatic activity of components of the mitochondrial electron transport chain [103]. Whether such alterations result in global effects on mitochondrial function remains unclear [104], but the potential effects of past and present ART should be considered as older adults with HIV infection are maintained on lifelong therapy. These and other parallels in aging and HIV infection are summarized in Table 1 and Figure 1.

Table 1.

Comparison of the alterations in innate immunity and signs of immunosenescence seen in HIV infection and Aging.

HIV Infection Aging Immune Category
Heightened pro-inflammatory environment;
  • Increased IL-6, TNF-a, Neopterin, sCD14 [69].

  • Increased coagulation factors and acute phase reactants [69].

Inflamm-Aging;
  • Increased cytokines, neopterin [1112].

  • Increased coagulation factors, acute phase reactants [1112].

Pro-inflammatory environment; signs of immune activation
  • Inflammatory CD14+, CD16+ monocytes are increased [15].

  • Monocytes of young HIV infected subjects show increased expression of CD11b, and decreased expression of CD62L and CD115 [15].

  • Plasma levels of soluble CD163 and CXCL10 resemble those seen in older adults [15,19].

  • Exhibit impaired phagocytic function, and telomere shortening [15].

  • Inflammatory CD14+, CD16+ monocytes are increased [16–18].

  • Increased expression of CD11b [16].

  • Decreased expression of CD62L and CD115 [16].

  • Plasma levels of soluble CD163 and CXCL10 increase with age [16].

  • Exhibit impaired phagocytosis and shortening of telomeres [16].

Monocytes
  • TLR7/8 and TLR2 cytokine production is increased in HIV infection [23].

  • Increased expression of TLR2, which is upregulated with exposure to gp120 [24].

  • Decrease in TLR1/2 mediated cytokine production, and TLR1 surface expression [18,20].

  • Increased TLR4 induced cytokine production in CD14+ CD16+ monocytes [16].

  • Increased TLR5 induced cytokine production (adherent monocytes) [22].

Monocytes/TLRs
  • TLR-8 induced TNF-a is decreased in mDC at the earliest stages of primary infection [34], increased in later stages of primary infection [23,35].

  • TLR9 induced cytokine production is decreased at all stages of infection in pDC [23].

  • Increased plasma levels of IFN-α [48].

  • Diminished numbers of pDCs in peripheral blood [41,42].

  • Overall decreased IFN-a production in pDC [4446].

  • In chronic HIV infection, decreased TLR-dependent IL-12 production is reported in monocyte-derived DCs [3739]

  • Overall decreased TLR-dependent cytokine production in both mDC and pDC [2530].

  • Increased basal cytokine production [26].

  • Diminished numbers of pDCs noted in the peripheral blood [27,43].

  • Decreased IFN-a production by pDC [26, 27, 29, 30, 43].

  • Monocyte-derived DCs show increased cytokine production in response to TLR4 and 8 induction and treatment with self-DNA [31,32]

  • Monocyte-derived DCs have decreased type I interferon production in response to West Nile virus [33]

Dendritic cells and TLRs
  • HIV viremia; HIV RNA stimulates TLR 7/8 [5052]

  • gp120 stimulates TLR 2 and 4 in genital epithelial cells [57].

  • Co-infection with herpesviruses; CMV [71].

  • Intestinal mucosa compromise and subsequent microbial translocation [58,59].

  • CMV reactivation is increased in older adults [68].

  • Increased plasma LPS binding protein (surrogate for Microbial translocation) [66].

PAMPs
  • Increased cell death and pyroptosis of CD4 cells [80].

  • Increased plasma cell free DNA [7476].

DAMPs
  • Hypogonadism and low testosterone are common among HIV infected men [87].

  • Fat redistribution occurs with lipodystrophy, a side effect of ART, and is associated withincreased pro-inflammatory cytokines [95].

  • ART causes inhibition of mitochondrial DNA polymerase γ and leads to increased mtDNA mutations and loss of mtDNA [9699].

  • Decreases in sex hormone production are associated with elevated pro-inflammatory cytokine levels [8186].

  • Redistricution of adipose tissue [88].

  • Adipose tissue is a source of pro-inflammatory cytokines, and increases with age [8994].

  • mtDNA mutations accumulate with age [102].

Hormones/Metabolic Factors
  • NRTIs inhibittelomerase activity in vitro, and are associated with shortening of telomeres [106, 107].

  • Replicative senescence in aging is accompanied by telomere shortening [109].

Senescence

Figure 1.

Figure 1

Comparison of effects of HIV and aging on innate immune responses. Both HIV and Aging are accompanied by pro-inflammatory environments that are likely to be triggered by elevated levels of pattern recognition ligands (Pathogen-Associated Molecular patterns (PAMPS) and Damage-Associated Molecular Patterns (DAMPS)). Toll-like receptors (TLR) and IFN-α receptor (IFNAR) are depicted. Increases or decreases in TLR-induced cytokine responses are shown by arrows (blue arrow: DC, Orange arrow: Monocytes, Green arrow; Monocyte-derived DC). Abbreviations: IFN, Interferon; mtDNA, mitochondrial DNA; TERT, Telomerase reverse transcriptase; NRTIs, nucleotide reverse transcriptase inhibitors; LPS, Lipopolysaccharide.

Inline graphic TLR8 induced TNF-α is decreased in mDC at the earliest stages of infection

Conclusion

With effective ART, HIV infection is now manageable as a chronic illness. For patients surviving into old age, geriatric issues of multiple co-morbidities and polypharmacy become crucial issues in management, and important covariates in immunologic analyses of such individuals. Moreover, factors such as the duration of HIV infection, and duration, efficacy, and adverse effects of ART will influence inflammatory pathways and age-associated pathways such as telomerase. In this regard, a reasonable hypothetical question would compare innate immune activation in a 30-year-old infected with HIV since birth to a newly infected 60-year-old—and whether merely classifying these subjects on the basis of chronologic age without considering duration of infection and treatment would be appropriate. Future studies of aging adults with HIV infection will need to embrace this complexity.

Text Box.

Effects of ART on Telomerase

The Telomerase Reverse Transcriptase (TERT) shares homology to retroviral reverse transcriptases [105], including that of HIV-1. NRTIs inhibit telomerase activity in vitro, and are associated with telomere shortening [106,107]. Telomerase inhibition was observed not only for older generation NRTIs, but also for widely used drugs, including Tenofovir (the most potent telomerase inhibitor in one study [106] and Abacavir. In addition, a recent report suggests that the Vpr protein of HIV-1 may also decrease telomerase activity by enhancing ubiquitination of TERT [108]. The juxtaposition of age-associated telomere shortening, in part reflecting replicative senescence [109], and potential long-term effects of ART may further enhance aging phenotypes in the immune system of older adults with HIV infection.

Highlights.

  • A dysregulated pro-inflammatory state is present in both aging and HIV infection.

  • Age- and HIV-related changes in TLR function may contribute to this dysregulation.

  • DNA DAMPs, other viruses, fat and hormonal changes also result in dysregulation.

  • Effects of microbial translocation and antiretroviral drugs may accumulate with age.

  • HIV disease and therapy duration and comorbid illnesses complicate studies of aging.

Acknowledgements

The authors’ work has been supported by grants from the US National Institutes of Health, National Institute of Allergy and Infectious Diseases T32 AI007517 (to H.J.Z.), U19 AI089992, and contract 272201100019C-3-0-1 (to A.C.S.), and by National Institute on Aging grant K24 AG042489 (to A.C.S.).

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.

Contributor Information

Heidi J. Zapata, Email: heidi.zapata@yale.edu.

Albert C. Shaw, Email: albert.shaw@yale.edu.

References

  • 1.Samji H, Cescon A, Hogg RS, Modur SP, Althoff KN, Buchacz K, Burchell AN, Cohen M, Gebo KA, Gill MJ, et al. Closing the Gap: Increases in Life Expectancy among Treated HIV-Positive Individuals in the United States and Canada. PLoS One. 2013;8:e81355. doi: 10.1371/journal.pone.0081355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Antiretroviral Therapy Cohort C. Life expectancy of individuals on combination antiretroviral therapy in high-income countries: a collaborative analysis of 14 cohort studies. Lancet. 2008;372:293–299. doi: 10.1016/S0140-6736(08)61113-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Luther VP, Wilkin AM. HIV infection in older adults. Clin Geriatr Med. 2007;23:567–583. doi: 10.1016/j.cger.2007.02.004. vii. [DOI] [PubMed] [Google Scholar]
  • 4.High KP, Brennan-Ing M, Clifford DB, Cohen MH, Currier J, Deeks SG, Deren S, Effros RB, Gebo K, Goronzy JJ, et al. HIV and aging: state of knowledge and areas of critical need for research. A report to the NIH Office of AIDS Research by the HIV and Aging Working Group. J Acquir Immune Defic Syndr. 2012;60(Suppl 1):S1–S18. doi: 10.1097/QAI.0b013e31825a3668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Effros RB, Fletcher CV, Gebo K, Halter JB, Hazzard WR, Horne FM, Huebner RE, Janoff EN, Justice AC, Kuritzkes D, et al. Aging and infectious diseases: workshop on HIV infection and aging: what is known and future research directions. Clin Infect Dis. 2008;47:542–553. doi: 10.1086/590150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Keating SM, Jacobs ES, Norris PJ. Soluble mediators of inflammation in HIV and their implications for therapeutics and vaccine development. Cytokine Growth Factor Rev. 2012;23:193–206. doi: 10.1016/j.cytogfr.2012.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Armah KA, McGinnis K, Baker J, Gibert C, Butt AA, Bryant KJ, Goetz M, Tracy R, Oursler KK, Rimland D, et al. HIV status, burden of comorbid disease, and biomarkers of inflammation, altered coagulation, and monocyte activation. Clin Infect Dis. 2012;55:126–136. doi: 10.1093/cid/cis406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, Ledergerber B, Lundgren J, Neuhaus J, Nixon D, 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]
  • 9.Kamat A, Misra V, Cassol E, Ancuta P, Yan Z, Li C, Morgello S, Gabuzda D. A plasma biomarker signature of immune activation in HIV patients on antiretroviral therapy. PLoS One. 2012;7:e30881. doi: 10.1371/journal.pone.0030881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Krishnan S, Wilson EM, Sheikh V, Rupert A, Mendoza D, Yang J, Lempicki R, Migueles SA, Sereti I. Evidence for Innate Immune System Activation in HIV Type 1-Infected Elite Controllers. J Infect Dis. 2014;209:931–939. doi: 10.1093/infdis/jit581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Leng SX, Tian X, Matteini A, Li H, Hughes J, Jain A, Walston JD, Fedarko NS. IL-6-independent association of elevated serum neopterin levels with prevalent frailty in community-dwelling older adults. Age Ageing. 2011;40:475–481. doi: 10.1093/ageing/afr047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol. 2013;13:875–887. doi: 10.1038/nri3547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Salvioli S, Capri M, Valensin S, Tieri P, Monti D, Ottaviani E, Franceschi C. Inflamm-aging, cytokines and aging: state of the art, new hypotheses on the role of mitochondria and new perspectives from systems biology. Curr Pharm Des. 2006;12:3161–3171. doi: 10.2174/138161206777947470. [DOI] [PubMed] [Google Scholar]
  • 14.Franceschi C, Valensin S, Bonafe M, Paolisso G, Yashin AI, Monti D, De Benedictis G. The network and the remodeling theories of aging: historical background and new perspectives. Exp Gerontol. 2000;35:879–896. doi: 10.1016/s0531-5565(00)00172-8. [DOI] [PubMed] [Google Scholar]
  • 15. Hearps AC, Maisa A, Cheng WJ, Angelovich TA, Lichtfuss GF, Palmer CS, Landay AL, Jaworowski A, Crowe SM. HIV infection induces age-related changes to monocytes and innate immune activation in young men that persist despite combination antiretroviral therapy. AIDS. 2012;26:843–853. doi: 10.1097/QAD.0b013e328351f756. *A study concluding that indices of monocyte activation in younger HIV-infected men resembled those in elderly HIV-negative controls. Monocytes from young HIV-infected men also had impaired phagocytosis and shortened telomere length compared to age-matched HIV-negative subjects.
  • 16.Hearps AC, Martin GE, Angelovich TA, Cheng WJ, Maisa A, Landay AL, Jaworowski A, Crowe SM. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell. 2012;11:867–875. doi: 10.1111/j.1474-9726.2012.00851.x. [DOI] [PubMed] [Google Scholar]
  • 17.Seidler S, Zimmermann HW, Bartneck M, Trautwein C, Tacke F. Age-dependent alterations of monocyte subsets and monocyte-related chemokine pathways in healthy adults. BMC Immunol. 2010;11:30. doi: 10.1186/1471-2172-11-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nyugen J, Agrawal S, Gollapudi S, Gupta S. Impaired functions of peripheral blood monocyte subpopulations in aged humans. J Clin Immunol. 2010;30:806–813. doi: 10.1007/s10875-010-9448-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Martin GE, Gouillou M, Hearps AC, Angelovich TA, Cheng AC, Lynch F, Cheng WJ, Paukovics G, Palmer CS, Novak RM, et al. Age-associated changes in monocyte and innate immune activation markers occur more rapidly in HIV infected women. PLoS One. 2013;8:e55279. doi: 10.1371/journal.pone.0055279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.van Duin D, Mohanty S, Thomas V, Ginter S, Montgomery RR, Fikrig E, Allore HG, Medzhitov R, Shaw AC. Age-associated defect in human TLR-1/2 function. J Immunol. 2007;178:970–975. doi: 10.4049/jimmunol.178.2.970. [DOI] [PubMed] [Google Scholar]
  • 21.van Duin D, Allore HG, Mohanty S, Ginter S, Newman FK, Belshe RB, Medzhitov R, Shaw AC. Prevaccine determination of the expression of costimulatory B7 molecules in activated monocytes predicts influenza vaccine responses in young and older adults. J Infect Dis. 2007;195:1590–1597. doi: 10.1086/516788. [DOI] [PubMed] [Google Scholar]
  • 22.Qian F, Wang X, Zhang L, Chen S, Piecychna M, Allore H, Bockenstedt L, Malawista S, Bucala R, Shaw AC, et al. Age-associated elevation in TLR5 leads to increased inflammatory responses in the elderly. Aging Cell. 2012;11:104–110. doi: 10.1111/j.1474-9726.2011.00759.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chang JJ, Lacas A, Lindsay RJ, Doyle EH, Axten KL, Pereyra F, Rosenberg ES, Walker BD, Allen TM, Altfeld M. Differential regulation of toll-like receptor pathways in acute and chronic HIV-1 infection. AIDS. 2012;26:533–541. doi: 10.1097/QAD.0b013e32834f3167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Heggelund L, Muller F, Lien E, Yndestad A, Ueland T, Kristiansen KI, Espevik T, Aukrust P, Froland SS. Increased expression of toll-like receptor 2 on monocytes in HIV infection: possible roles in inflammation and viral replication. Clin Infect Dis. 2004;39:264–269. doi: 10.1086/421780. [DOI] [PubMed] [Google Scholar]
  • 25.Della Bella S, Bierti L, Presicce P, Arienti R, Valenti M, Saresella M, Vergani C, Villa ML. Peripheral blood dendritic cells and monocytes are differently regulated in the elderly. Clin Immunol. 2007;122:220–228. doi: 10.1016/j.clim.2006.09.012. [DOI] [PubMed] [Google Scholar]
  • 26.Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, Chen S, Towle V, Belshe RB, Fikrig E, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol. 2010;184:2518–2527. doi: 10.4049/jimmunol.0901022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum Immunol. 2009;70:777–784. doi: 10.1016/j.humimm.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Garbe K, Bratke K, Wagner S, Virchow JC, Lommatzsch M. Plasmacytoid dendritic cells and their Toll-like receptor 9 expression selectively decrease with age. Hum Immunol. 2012;73:493–497. doi: 10.1016/j.humimm.2012.02.007. [DOI] [PubMed] [Google Scholar]
  • 29.Canaday DH, Amponsah NA, Jones L, Tisch DJ, Hornick TR, Ramachandra L. Influenza-induced production of interferon-alpha is defective in geriatric individuals. J Clin Immunol. 2010;30:373–383. doi: 10.1007/s10875-010-9374-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sridharan A, Esposo M, Kaushal K, Tay J, Osann K, Agrawal S, Gupta S, Agrawal A. Age-associated impaired plasmacytoid dendritic cell functions lead to decreased CD4 and CD8 T cell immunity. Age (Dordr) 2011;33:363–376. doi: 10.1007/s11357-010-9191-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Agrawal A, Agrawal S, Cao JN, Su H, Osann K, Gupta S. Altered innate immune functioning of dendritic cells in elderly humans: a role of phosphoinositide 3-kinase-signaling pathway. J Immunol. 2007;178:6912–6922. doi: 10.4049/jimmunol.178.11.6912. [DOI] [PubMed] [Google Scholar]
  • 32.Agrawal A, Tay J, Ton S, Agrawal S, Gupta S. Increased reactivity of dendritic cells from aged subjects to self-antigen, the human DNA. J Immunol. 2009;182:1138–1145. doi: 10.4049/jimmunol.182.2.1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Qian F, Wang X, Zhang L, Lin A, Zhao H, Fikrig E, Montgomery RR. Impaired interferon signaling in dendritic cells from older donors infected in vitro with West Nile virus. J Infect Dis. 2011;203:1415–1424. doi: 10.1093/infdis/jir048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Frleta D, Ochoa CE, Kramer HB, Khan SA, Stacey AR, Borrow P, Kessler BM, Haynes BF, Bhardwaj N. HIV-1 infection-induced apoptotic microparticles inhibit human DCs via CD44. J Clin Invest. 2012;122:4685–4697. doi: 10.1172/JCI64439. **Myeloid DCs from subjects with early primary HIV-1 infection had impaired TLR8-dependent cytokine production compared to subjects at later stages of primary infection of HIV-negative controls; plasma from subjects with primary infection could induce this inhibition, which was mediated by apoptotic microparticles released during acute HIV infection.
  • 35.Sabado RL, O’Brien M, Subedi A, Qin L, Hu N, Taylor E, Dibben O, Stacey A, Fellay J, Shianna KV, et al. Evidence of dysregulation of dendritic cells in primary HIV infection. Blood. 2010;116:3839–3852. doi: 10.1182/blood-2010-03-273763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Huang J, Yang Y, Al-Mozaini M, Burke PS, Beamon J, Carrington MF, Seiss K, Rychert J, Rosenberg ES, Lichterfeld M, et al. Dendritic cell dysfunction during primary HIV-1 infection. J Infect Dis. 2011;204:1557–1562. doi: 10.1093/infdis/jir616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Miller EA, Spadaccia MR, O’Brien MP, Rolnitzky L, Sabado R, Manches O, Frleta D, Bhardwaj N. Plasma factors during chronic HIV-1 infection impair IL-12 secretion by myeloid dendritic cells via a virus-independent pathway. J Acquir Immune Defic Syndr. 2012;61:535–544. doi: 10.1097/QAI.0b013e31826afbce. *Subjects with chronic untreated HIV infection demonstrated impaired TLR3-induced cytokine production compared to HIV-negative controls; inhibition was partially improved with ART.
  • 38.Buisson S, Benlahrech A, Gazzard B, Gotch F, Kelleher P, Patterson S. Monocyte- derived dendritic cells from HIV type 1-infected individuals show reduced ability to stimulate T cells and have altered production of interleukin (IL)-12 and IL-10. J Infect Dis. 2009;199:1862–1871. doi: 10.1086/599122. [DOI] [PubMed] [Google Scholar]
  • 39.Fan Z, Huang XL, Kalinski P, Young S, Rinaldo CR., Jr. Dendritic cell function during chronic hepatitis C virus and human immunodeficiency virus type 1 infection. Clin Vaccine Immunol. 2007;14:1127–1137. doi: 10.1128/CVI.00141-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Blanchet FP, Moris A, Nikolic DS, Lehmann M, Cardinaud S, Stalder R, Garcia E, Dinkins C, Leuba F, Wu L, et al. Human immunodeficiency virus-1 inhibition of immunoamphisomes in dendritic cells impairs early innate and adaptive immune responses. Immunity. 2010;32:654–669. doi: 10.1016/j.immuni.2010.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Lehmann C, Jung N, Forster K, Koch N, Leifeld L, Fischer J, Mauss S, Drebber U, Steffen HM, Romerio F, et al. Longitudinal Analysis of Distribution and Function of Plasmacytoid Dendritic Cells in Peripheral Blood and Gut Mucosa of HIV Infected Patients. J Infect Dis. 2014;209:940–949. doi: 10.1093/infdis/jit612. *A longitudinal analysis of HIV-infected subjects at baseline and 6 months after initiation of ART that evaluated pDCs in blood and from ileal biopsies. The proportion of pDCs in gut-associated lymphoid tissue was increased in HIV-infected subjects prior to starting ART, correlating with upregulation of the gut homing receptor CD103.
  • 42.Lehmann C, Lafferty M, Garzino-Demo A, Jung N, Hartmann P, Fatkenheuer G, Wolf JS, van Lunzen J, Romerio F. Plasmacytoid dendritic cells accumulate and secrete interferon alpha in lymph nodes of HIV-1 patients. PLoS One. 2010;5:e11110. doi: 10.1371/journal.pone.0011110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shodell M, Siegal FP. Circulating, interferon-producing plasmacytoid dendritic cells decline during human ageing. Scand J Immunol. 2002;56:518–521. doi: 10.1046/j.1365-3083.2002.01148.x. [DOI] [PubMed] [Google Scholar]
  • 44.Kamga I, Kahi S, Develioglu L, Lichtner M, Maranon C, Deveau C, Meyer L, Goujard C, Lebon P, Sinet M, et al. Type I interferon production is profoundly and transiently impaired in primary HIV-1 infection. J Infect Dis. 2005;192:303–310. doi: 10.1086/430931. [DOI] [PubMed] [Google Scholar]
  • 45.Tilton JC, Manion MM, Luskin MR, Johnson AJ, Patamawenu AA, Hallahan CW, Cogliano-Shutta NA, Mican JM, Davey RT, Jr., Kottilil S, et al. Human immunodeficiency virus viremia induces plasmacytoid dendritic cell activation in vivo and diminished alpha interferon production in vitro. J Virol. 2008;82:3997–4006. doi: 10.1128/JVI.01545-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Reitano KN, Kottilil S, Gille CM, Zhang X, Yan M, O’Shea MA, Roby G, Hallahan CW, Yang J, Lempicki RA, et al. Defective plasmacytoid dendritic cell-NK cell cross-talk in HIV infection. AIDS Res Hum Retroviruses. 2009;25:1029–1037. doi: 10.1089/aid.2008.0311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Martinelli E, Cicala C, Van Ryk D, Goode DJ, Macleod K, Arthos J, Fauci AS. HIV-1 gp120 inhibits TLR9-mediated activation and IFN-{alpha} secretion in plasmacytoid dendritic cells. Proc Natl Acad Sci U S A. 2007;104:3396–3401. doi: 10.1073/pnas.0611353104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.von Sydow M, Sonnerborg A, Gaines H, Strannegard O. Interferon-alpha and tumor necrosis factor-alpha in serum of patients in various stages of HIV-1 infection. AIDS Res Hum Retroviruses. 1991;7:375–380. doi: 10.1089/aid.1991.7.375. [DOI] [PubMed] [Google Scholar]
  • 49.O’Brien M, Manches O, Sabado RL, Baranda SJ, Wang Y, Marie I, Rolnitzky L, Markowitz M, Margolis DM, Levy D, et al. Spatiotemporal trafficking of HIV in human plasmacytoid dendritic cells defines a persistently IFN-alpha-producing and partially matured phenotype. J Clin Invest. 2011;121:1088–1101. doi: 10.1172/JCI44960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Beignon AS, McKenna K, Skoberne M, Manches O, DaSilva I, Kavanagh DG, Larsson M, Gorelick RJ, Lifson JD, Bhardwaj N. Endocytosis of HIV-1 activates plasmacytoid dendritic cells via Toll-like receptor-viral RNA interactions. J Clin Invest. 2005;115:3265–3275. doi: 10.1172/JCI26032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Meier A, Alter G, Frahm N, Sidhu H, Li B, Bagchi A, Teigen N, Streeck H, Stellbrink HJ, Hellman J, et al. MyD88-dependent immune activation mediated by human immunodeficiency virus type 1-encoded Toll-like receptor ligands. J Virol. 2007;81:8180–8191. doi: 10.1128/JVI.00421-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004;303:1526–1529. doi: 10.1126/science.1093620. [DOI] [PubMed] [Google Scholar]
  • 53.Hatano H, Strain MC, Scherzer R, Bacchetti P, Wentworth D, Hoh R, Martin JN, McCune JM, Neaton JD, Tracy RP, et al. Increase in 2-long terminal repeat circles and decrease in D-dimer after raltegravir intensification in patients with treated HIV infection: a randomized, placebo-controlled trial. J Infect Dis. 2013;208:1436–1442. doi: 10.1093/infdis/jit453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chun TW, Fauci AS. Viral persistence in HIV infection: much known, much to learn. J Infect Dis. 2013;208:1356–1358. doi: 10.1093/infdis/jit455. [DOI] [PubMed] [Google Scholar]
  • 55.Mureith MW, Chang JJ, Lifson JD, Ndung’u T, Altfeld M. Exposure to HIV-1- encoded Toll-like receptor 8 ligands enhances monocyte response to microbial encoded Toll-like receptor 2/4 ligands. AIDS. 2010;24:1841–1848. doi: 10.1097/QAD.0b013e32833ad89a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lester RT, Yao XD, Ball TB, McKinnon LR, Omange WR, Kaul R, Wachihi C, Jaoko W, Rosenthal KL, Plummer FA. 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]
  • 57.Nazli A, Kafka JK, Ferreira VH, Anipindi V, Mueller K, Osborne BJ, Dizzell S, Chauvin S, Mian MF, Ouellet M, et al. HIV-1 gp120 induces TLR2- and TLR4-mediated innate immune activation in human female genital epithelium. J Immunol. 2013;191:4246–4258. doi: 10.4049/jimmunol.1301482. [DOI] [PubMed] [Google Scholar]
  • 58.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]
  • 59.Sandler NG, Douek DC. Microbial translocation in HIV infection: causes, consequences and treatment opportunities. Nat Rev Microbiol. 2012;10:655–666. doi: 10.1038/nrmicro2848. [DOI] [PubMed] [Google Scholar]
  • 60.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, 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]
  • 61.Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013;21:6–13. doi: 10.1016/j.tim.2012.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, Landay A, Martin J, Sinclair E, Asher AI, 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]
  • 63.Merlini E, Bai F, Bellistri GM, Tincati C, d’Arminio Monforte A, Marchetti G. 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]
  • 64.Ellis CL, Ma ZM, Mann SK, Li CS, Wu J, Knight TH, Yotter T, Hayes TL, Maniar AH, Troia-Cancio PV, 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]
  • 65.Dillon SM, Lee EJ, Kotter CV, Austin GL, Dong Z, Hecht DK, Gianella S, Siewe B, Smith DM, Landay AL, et al. An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal Immunol. 2014 doi: 10.1038/mi.2013.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Stehle JR, Jr., Leng X, Kitzman DW, Nicklas BJ, Kritchevsky SB, High KP. Lipopolysaccharide-binding protein, a surrogate marker of microbial translocation, is associated with physical function in healthy older adults. J Gerontol A Biol Sci Med Sci. 2012;67:1212–1218. doi: 10.1093/gerona/gls178. *The first study to show an association between a surrogate marker of microbial translocation and measurements of physical function in HIV-negative older adults (mean age 69).
  • 67.Limaye AP, Kirby KA, Rubenfeld GD, Leisenring WM, Bulger EM, Neff MJ, Gibran NS, Huang ML, Santo Hayes TK, Corey L, et al. Cytomegalovirus reactivation in critically ill immunocompetent patients. JAMA. 2008;300:413–422. doi: 10.1001/jama.300.4.413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stowe RP, Kozlova EV, Yetman DL, Walling DM, Goodwin JS, Glaser R. Chronic herpesvirus reactivation occurs in aging. Exp Gerontol. 2007;42:563–570. doi: 10.1016/j.exger.2007.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Roberts ET, Haan MN, Dowd JB, Aiello AE. Cytomegalovirus antibody levels, inflammation, and mortality among elderly Latinos over 9 years of follow-up. Am J Epidemiol. 2010;172:363–371. doi: 10.1093/aje/kwq177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Bartlett DB, Firth CM, Phillips AC, Moss P, Baylis D, Syddall H, Sayer AA, Cooper C, Lord JM. The age-related increase in low-grade systemic inflammation (Inflammaging) is not driven by cytomegalovirus infection. Aging Cell. 2012;11:912–915. doi: 10.1111/j.1474-9726.2012.00849.x. *A longitudinal study that followed adults who were CMV seropositive, CMV seronegative, and those who seroconverted to CMV over a 10-year period. The study showed no differences in changes in cytokine levels among these groups.
  • 71.Deayton JR, Prof Sabin CA, Johnson MA, Emery VC, Wilson P, Griffiths PD. Importance of cytomegalovirus viraemia in risk of disease progression and death in HIV-infected patients receiving highly active antiretroviral therapy. Lancet. 2004;363:2116–2121. doi: 10.1016/S0140-6736(04)16500-8. [DOI] [PubMed] [Google Scholar]
  • 72.Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118. doi: 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6:2853–2868. doi: 10.1371/journal.pbio.0060301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Jylhava J, Jylha M, Lehtimaki T, Hervonen A, Hurme M. Circulating cell-free DNA is associated with mortality and inflammatory markers in nonagenarians: the Vitality 90+ Study. Exp Gerontol. 2012;47:372–378. doi: 10.1016/j.exger.2012.02.011. [DOI] [PubMed] [Google Scholar]
  • 75. Jylhava J, Nevalainen T, Marttila S, Jylha M, Hervonen A, Hurme M. Characterization of the role of distinct plasma cell-free DNA species in age-associated inflammation and frailty. Aging Cell. 2013;12:388–397. doi: 10.1111/acel.12058. *In a nonagenarian cohort compared to young controls, total and unmethylated cell-free DNA in plasma was associated with the geriatric syndrome of frailty and transcriptional activation of pro-inflammatory pathways (see also reference 83 from the same group).
  • 76. Pinti M, Cevenini E, Nasi M, De Biasi S, Salvioli S, Monti D, Benatti S, Gibellini L, Cotichini R, Stazi MA, et al. Circulating mitochondrial DNA increases with age and is a familiar trait: implications for “inflamm-aging”. Eur J Immunol. 2014;44:1552–1562. doi: 10.1002/eji.201343921. *Evaluation of circulating mitochondrial DNA (mtDNA) in plasma samples from 831 subjects ranging in age from 1–104 showed that mtDNA levels increase after the fifth decade, and that increased plasma mtDNA correlated with increased pro-inflammatory cytokine levels.
  • 77.Iyer SS, Pulskens WP, Sadler JJ, Butter LM, Teske GJ, Ulland TK, Eisenbarth SC, Florquin S, Flavell RA, Leemans JC, et al. Necrotic cells trigger a sterile inflammatory response through the Nlrp3 inflammasome. Proc Natl Acad Sci U S A. 2009;106:20388–20393. doi: 10.1073/pnas.0908698106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Li H, Ambade A, Re F. Cutting edge: Necrosis activates the NLRP3 inflammasome. J Immunol. 2009;183:1528–1532. doi: 10.4049/jimmunol.0901080. [DOI] [PubMed] [Google Scholar]
  • 79.Imaeda AB, Watanabe A, Sohail MA, Mahmood S, Mohamadnejad M, Sutterwala FS, Flavell RA, Mehal WZ. Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9 and the Nalp3 inflammasome. J Clin Invest. 2009;119:305–314. doi: 10.1172/JCI35958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Doitsh G, Galloway NL, Geng X, Yang Z, Monroe KM, Zepeda O, Hunt PW, Hatano H, Sowinski S, Munoz-Arias I, et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature. 2014;505:509–514. doi: 10.1038/nature12940. **Using ex vivo human lymphoid aggregate tissue culture, and flourescently labeled caspase-1 and caspase-3, this study showed that during HIV infection non-productively infected CD4 T cells die via caspase -1-mediated pyroptosis.
  • 81.Ray A, Prefontaine KE, Ray P. Down-modulation of interleukin-6 gene expression by 17 beta-estradiol in the absence of high affinity DNA binding by the estrogen receptor. J Biol Chem. 1994;269:12940–12946. [PubMed] [Google Scholar]
  • 82.Pottratz ST, Bellido T, Mocharla H, Crabb D, Manolagas SC. 17 beta-Estradiol inhibits expression of human interleukin-6 promoter-reporter constructs by a receptor-dependent mechanism. J Clin Invest. 1994;93:944–950. doi: 10.1172/JCI117100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Abu-Taha M, Rius C, Hermenegildo C, Noguera I, Cerda-Nicolas JM, Issekutz AC, Jose PJ, Cortijo J, Morcillo EJ, Sanz MJ. Menopause and ovariectomy cause a low grade of systemic inflammation that may be prevented by chronic treatment with low doses of estrogen or losartan. J Immunol. 2009;183:1393–1402. doi: 10.4049/jimmunol.0803157. [DOI] [PubMed] [Google Scholar]
  • 84.Gordon CM, LeBoff MS, Glowacki J. Adrenal and gonadal steroids inhibit IL-6 secretion by human marrow cells. Cytokine. 2001;16:178–186. doi: 10.1006/cyto.2001.0962. [DOI] [PubMed] [Google Scholar]
  • 85.Maggio M, Basaria S, Ble A, Lauretani F, Bandinelli S, Ceda GP, Valenti G, Ling SM, Ferrucci L. Correlation between testosterone and the inflammatory marker soluble interleukin-6 receptor in older men. J Clin Endocrinol Metab. 2006;91:345–347. doi: 10.1210/jc.2005-1097. [DOI] [PubMed] [Google Scholar]
  • 86.Yang BC, Liu CW, Chen YC, Yu CK. Exogenous dehydroepiandrosterone modified the expression of T helper-related cytokines in NZB/NZW F1 mice. Immunol Invest. 1998;27:291–302. doi: 10.3109/08820139809070902. [DOI] [PubMed] [Google Scholar]
  • 87.Monroe AK, Dobs AS, Palella FJ, Kingsley LA, Witt MD, Brown TT. Morning free and total testosterone in HIV-infected men: implications for the assessment of hypogonadism. AIDS Res Ther. 2014;11:6. doi: 10.1186/1742-6405-11-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, Khosla S, Jensen MD, Kirkland JL. Fat tissue, aging, and cellular senescence. Aging Cell. 2010;9:667–684. doi: 10.1111/j.1474-9726.2010.00608.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Morin CL, Pagliassotti MJ, Windmiller D, Eckel RH. Adipose tissue-derived tumor necrosis factor-alpha activity is elevated in older rats. J Gerontol A Biol Sci Med Sci. 1997;52:B190–B195. doi: 10.1093/gerona/52a.4.b190. [DOI] [PubMed] [Google Scholar]
  • 90.Wu D, Ren Z, Pae M, Guo W, Cui X, Merrill AH, Meydani SN. Aging up-regulates expression of inflammatory mediators in mouse adipose tissue. J Immunol. 2007;179:4829–4839. doi: 10.4049/jimmunol.179.7.4829. [DOI] [PubMed] [Google Scholar]
  • 91.Starr ME, Evers BM, Saito H. Age-associated increase in cytokine production during systemic inflammation: adipose tissue as a major source of IL-6. J Gerontol A Biol Sci Med Sci. 2009;64:723–730. doi: 10.1093/gerona/glp046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cartier A, Cote M, Lemieux I, Perusse L, Tremblay A, Bouchard C, Despres JP. Age- related differences in inflammatory markers in men: contribution of visceral adiposity. Metabolism. 2009;58:1452–1458. doi: 10.1016/j.metabol.2009.04.025. [DOI] [PubMed] [Google Scholar]
  • 93.Stenholm S, Koster A, Alley DE, Visser M, Maggio M, Harris TB, Egan JM, Bandinelli S, Guralnik JM, Ferrucci L. Adipocytokines and the metabolic syndrome among older persons with and without obesity: the InCHIANTI study. Clin Endocrinol (Oxf) 2010;73:55–65. doi: 10.1111/j.1365-2265.2009.03742.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pou KM, Massaro JM, Hoffmann U, Vasan RS, Maurovich-Horvat P, Larson MG, Keaney JF, Jr., Meigs JB, Lipinska I, Kathiresan S, et al. Visceral and subcutaneous adipose tissue volumes are cross-sectionally related to markers of inflammation and oxidative stress: the Framingham Heart Study. Circulation. 2007;116:1234–1241. doi: 10.1161/CIRCULATIONAHA.107.710509. [DOI] [PubMed] [Google Scholar]
  • 95.Koethe JR, Hulgan T, Niswender K. Adipose tissue and immune function: a review of evidence relevant to HIV infection. J Infect Dis. 2013;208:1194–1201. doi: 10.1093/infdis/jit324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Walker UA, Bickel M, Lutke Volksbeck SI, Ketelsen UP, Schofer H, Setzer B, Venhoff N, Rickerts V, Staszewski S. Evidence of nucleoside analogue reverse transcriptase inhibitor--associated genetic and structural defects of mitochondria in adipose tissue of HIV-infected patients. J Acquir Immune Defic Syndr. 2002;29:117–121. doi: 10.1097/00042560-200202010-00002. [DOI] [PubMed] [Google Scholar]
  • 97.Morse CG, Voss JG, Rakocevic G, McLaughlin M, Vinton CL, Huber C, Hu X, Yang J, Huang da W, Logun C, et al. HIV infection and antiretroviral therapy have divergent effects on mitochondria in adipose tissue. J Infect Dis. 2012;205:1778–1787. doi: 10.1093/infdis/jis101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cherry CL, Gahan ME, McArthur JC, Lewin SR, Hoy JF, Wesselingh SL. Exposure to dideoxynucleosides is reflected in lowered mitochondrial DNA in subcutaneous fat. J Acquir Immune Defic Syndr. 2002;30:271–277. doi: 10.1097/00126334-200207010-00002. [DOI] [PubMed] [Google Scholar]
  • 99.Nolan D, Hammond E, Martin A, Taylor L, Herrmann S, McKinnon E, Metcalf C, Latham B, Mallal S. Mitochondrial DNA depletion and morphologic changes in adipocytes associated with nucleoside reverse transcriptase inhibitor therapy. AIDS. 2003;17:1329–1338. doi: 10.1097/00002030-200306130-00007. [DOI] [PubMed] [Google Scholar]
  • 100.Perez-Matute P, Perez-Martinez L, Blanco JR, Oteo JA. Role of mitochondria in HIV infection and associated metabolic disorders: focus on nonalcoholic fatty liver disease and lipodystrophy syndrome. Oxid Med Cell Longev. 2013;2013:493413. doi: 10.1155/2013/493413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Payne BA, Wilson IJ, Hateley CA, Horvath R, Santibanez-Koref M, Samuels DC, Price DA, Chinnery PF. Mitochondrial aging is accelerated by anti-retroviral therapy through the clonal expansion of mtDNA mutations. Nat Genet. 2011;43:806–810. doi: 10.1038/ng.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bua E, Johnson J, Herbst A, Delong B, McKenzie D, Salamat S, Aiken JM. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. Am J Hum Genet. 2006;79:469–480. doi: 10.1086/507132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. McComsey GA, Daar ES, O’Riordan M, Collier AC, Kosmiski L, Santana JL, Fichtenbaum CJ, Fink H, Sax PE, Libutti DE, et al. Changes in fat mitochondrial DNA and function in subjects randomized to abacavir-lamivudine or tenofovir DF-emtricitabine with atazanavir-ritonavir or efavirenz: AIDS Clinical Trials Group study A5224s, substudy of A5202. J Infect Dis. 2013;207:604–611. doi: 10.1093/infdis/jis720. *Fat biopsies performed on HIV infected subjects randomized to ART regimens containing either ABC/3TC or TDF/FTC showed a decrease in mitochondrial DNA in both groups. Treatment with TDF/FTC was associated with mitochondrial respiratory chain dysfunction, as shown by decreased complex I and IV activity levels.
  • 104.Payne BA, Hollingsworth KG, Baxter J, Wilkins E, Lee V, Price DA, Trenell M, Chinnery PF. In vivo mitochondrial function in HIV-infected persons treated with contemporary anti-retroviral therapy: a magnetic resonance spectroscopy study. PLoS One. 2014;9:e84678. doi: 10.1371/journal.pone.0084678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Peng Y, Mian IS, Lue NF. Analysis of telomerase processivity: mechanistic similarity to HIV-1 reverse transcriptase and role in telomere maintenance. Mol Cell. 2001;7:1201–1211. doi: 10.1016/s1097-2765(01)00268-4. [DOI] [PubMed] [Google Scholar]
  • 106. Leeansyah E, Cameron PU, Solomon A, Tennakoon S, Velayudham P, Gouillou M, Spelman T, Hearps A, Fairley C, Smit de V, et al. Inhibition of telomerase activity by human immunodeficiency virus (HIV) nucleos(t)ide reverse transcriptase inhibitors: a potential factor contributing to HIV-associated accelerated aging. J Infect Dis. 2013;207:1157–1165. doi: 10.1093/infdis/jit006. *Activated PBMCs from HIV-infected subjects (on ART) were exposed to various NRTIs, which inhibited telomerase activity in vitro. Tenofovir was the most potent inhibitor of telomerase at therapeutic concentrations.
  • 107.Hukezalie KR, Thumati NR, Cote HC, Wong JM. In vitro and ex vivo inhibition of human telomerase by anti-HIV nucleoside reverse transcriptase inhibitors (NRTIs) but not by non-NRTIs. PLoS One. 2012;7:e47505. doi: 10.1371/journal.pone.0047505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wang X, Singh S, Jung HY, Yang G, Jun S, Sastry KJ, Park JI. HIV-1 Vpr protein inhibits telomerase activity via the EDD-DDB1-VPRBP E3 ligase complex. J Biol Chem. 2013;288:15474–15480. doi: 10.1074/jbc.M112.416735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dock JN, Effros RB. Role of CD8 T Cell Replicative Senescence in Human Aging and in HIV-mediated Immunosenescence. Aging Dis. 2011;2:382–397. [PMC free article] [PubMed] [Google Scholar]

RESOURCES