Abstract
More than one-third of adults in the United States are obese, and obesity-related disease accounts for some of the leading causes of preventable death. Mid-life obesity may be a strong predictor of physical function impairment later in life regardless of body mass index (BMI) in older age, highlighting the benefits of obesity prevention on health throughout the lifespan. Adipose tissue disturbances including lipodystrophy and obesity are prevalent in the setting of treated and untreated HIV infection. This article will review current knowledge on fat disturbances in HIV-infected persons, including therapeutic options and future directions.
Keywords: HIV, obesity, inflammation, adipose tissue, lipodystrophy, HIV/AIDS, HIV infection, obesity and HIV, BMI, antiretroviral therapy, review
Introduction
More than one-third of adults in the United States are obese, and obesity-related disease accounts for some of the leading causes of preventable death [1]. Longitudinal studies suggest that mid-life obesity may be a strong predictor of physical function impairment later in life regardless of body mass index (BMI) in older age [2, 3], highlighting the benefits of obesity prevention on health throughout the lifespan. Adipose tissue disturbances including lipodystrophy and generalized obesity are prevalent in the setting of treated and untreated HIV infection. This article will review current knowledge on fat disturbances in HIV-infected persons, including therapeutic options and future directions.
Discussion
Clinical Implications of Obesity and Body Fat Changes in HIV
The prevalence of obesity is increasing among HIV-infected persons, with up to 65% classified as overweight or obese [4-7]. ART initiation is often associated with weight gain partially attributed to a “return to health” phenomenon, with greater increases in weight seen in persons with the highest pre-ART HIV-1 RNA or lowest CD4+ T lymphocyte counts [8, 9]. In an AIDS Clinical Trial Group (ACTG) study (A5175) of ART initiation in resource-diverse settings, more than 25% of participants were overweight or obese at entry, and approximately 40% of participants were overweight or obese by week 144 [10]. These findings are consistent with prior observational studies on anthropomorphic changes in both resource-limited and resource-plentiful settings [11-13].
Weight gain following ART initiation has been associated with both beneficial and detrimental outcomes. While some weight gain may be beneficial particularly in wasted persons, there is likely a “tipping point” where weight gain begins to have a negative impact. For example, weight gain among underweight persons has been associated with a decline in high-sensitivity C-reactive protein (hs-CRP)levels, while weight gain among overweight or obese individuals has been associated with a significant increase in soluble CD14 (sCD14) levels (2015 Conference on Retroviruses and Opportunistic Infections, Seattle, WA; Erlandson, et al. Abstract 778). Similarly, a recent Veterans Aging Cohort Study analysis observed improved survival with weight gain in the first year of ART among underweight or normal weight but not overweight or obese participants [14]. Furthermore, overweight or obese HIV-infected individuals have a ≥67% prevalence of multi-morbidity [15]. In contrast, weight loss or failure to gain weight following ART initiation may be a poor prognostic sign or marker of concomitant infection or severe wasting [16].
Obesity is associated with hormone and cytokine imbalances, and chronic inflammation, all of which may contribute to its detrimental effects on multiple systems, including muscle [17]. Obesity and the metabolic syndrome are well-established risk factors for the development of physical function impairment or frailty among geriatric HIV-uninfected populations [18] and middle-aged or older HIV-infected adults [19-21]. Similarly, obesity is a known risk factor for diabetes mellitus and cardiovascular disease (CVD), and increased CVD risk has been described among obese and lipohypertrophic, HIV-infected adults [22].
While overall changes in body weight can have significant negative health impacts, the location of weight change may be of even greater importance. Adipose tissue accumulation in the trunk and viscera (lipohypertrophy) is well described following ART initiation. Up to 70% of HIV-infected individuals on ART may have abdominal or visceral obesity [8, 23-25], with changes not limited to older ART regimens. Indeed, in the recently completed ACTG study A5260, participants randomized to raltegravir or ritonavir-boosted darunavir or atazanavir with a backbone of tenofovir/emtricitabine had a mean visceral adipose tissue (VAT) gain of >30% after 96 weeks of therapy (2015 Conference on Retroviruses and Opportunistic Infections, Seattle, WA; McComsey, et al. Abstract 140). Discernment of lipohypertrophy in persons who are overweight or obese may be challenging, but a diagnosis of lipohypertrophy imparts important consequences and management considerations. In particular, changes in VAT of as little as 5% are believed to affect metabolic syndrome risk [26], and a large study of body composition and HIV in the US demonstrated that increased central fat was associated with greater five-year mortality [27].
In addition to metabolic and inflammatory consequences, body fat changes are stigmatizing and may impact self-esteem and overall health perception. These negative perceptions can impact ART adherence and quality of life [28-31]. In one study, two-thirds of HIV-infected participants reported that they would be willing to trade a year of life not to have lipodystrophy [32].
Finally, given associations between chronic inflammation and the development of comorbid disease [33-35], there is an urgent need to better understand the causes of and develop interventions to attenuate the effects of chronic inflammation and immune activation in people living with treated HIV infection, including addressing adipose tissue and obesity-associated inflammation.
How Adipose Tissue Contributes to Inflammation
In the setting of weight gain, adipocytes may increase in number (hyperplasia) or volume (hypertrophy). Large, hypertrophied adipocytes may become hypoxic or accumulate toxic ceramides or other lipids; hypertrophied adipocytes also recruit pro-inflammatory immune cells, primarily activated macrophages, and cell death may result [36-38]. Activated macrophages stimulate a type 1 immune response leading to the production of tumor necrosis factor (TNF)-α and interferon (IFN)-γ; activated macrophages also lose the ability to store iron, which is then deposited in adipose tissue and associated with reactive oxygen species production and mitochondrial dysfunction [39]. Ultimately, adipocyte hypertrophy is associated with higher systemic levels of pro-inflammatory cytokines including TNF-α, interleukin (IL)-6, IL-8, IFN-γ, and lower levels of the anti-inflammatory cytokine IL-10.
Imbalance in the production of adipokines (adiponectin and leptin) and infiltration of immune cells into adipose tissue exacerbate the pro-inflammatory environment. Adipocyte hypertrophy and VAT accumulation suppress adiponectin, stimulate transforming growth factor (TGF)-β production [40] and trigger pro-fibrotic processes. Pericellular fibrosis limits adipocyte size (particularly in omental adipose tissue) in an attempt to curb further adipose tissue expansion [41]. While adipose tissue fibrosis is a normal, compensatory response to fat gain, it has unintended consequences. First, adipose tissue fibrosis does not reverse with weight loss [41], making prevention key. Second, while fibrosis limits adipocyte hypertrophy, when overfeeding continues and adipocytes cannot expand, ectopic fat deposition into sites such as the viscera and skeletal muscle occur. Ectopic fat deposition is associated with additional inflammation and metabolic dysregulation. Awareness of adipose tissue as an inflammatory and endocrine organ has improved understanding of the role of adipose tissue dysfunction and the development of diseases of inflammation, including insulin resistance and CVD [42].
Data on adipose tissue disturbances in HIV-infected persons are limited and generally restricted to lipodystrophy rather than generalized obesity. Central lipohypertrophy is associated with increased adipose tissue inflammation and apoptosis [43], while dorsocervical lipohypertrophy is associated with adipose tissue fibrosis without inflammation, increased small adipocyte numbers and decreased vascularity [44]. A brown fat phenotype has been observed in dorsocervical subcutaneous adipose tissue (SAT) in association with protease inhibitor (PI) use and lipohypertrophy [44]. The shift to a brown fat phenotype, which is more metabolically active, may be an adaptive response to prevent further VAT expansion [45, 46], a hypothesis that is supported by the relative lack of inflammation in dorsocervical vs abdominal SAT in ART-treated patients irrespective of lipodystrophy status [47].
The known effects of specific antiretroviral agents on adipocytes are summarized in Table 1.
Table 1. Known effects of specific antiretroviral agents on adipocytes.
Type of Adipose Tissue Disturbance | Implicated Antiretroviral Agents |
---|---|
| |
Impaired adipogenesis or adipocyte differentiation or function | Lopinavir/ritonavir [48, 49], ritonavir [48], efavirenz [50, 51], rilpivirine [50], stavudine [52] |
Pre-adipocyte autophagy and apoptosis | Atazanavir [53] |
Impairment of lipid and/or glucose metabolism | Atazanavir/ritonavir [54], lopinavir/ritonavir [48, 49], ritonavir [48], ± darunavir [48, 49], NNRTIs [54], efavirenz [50], NRTI [54] |
Impairment of mitochondrial function | Atazanavir [53], azanavir/ritonavir [48], saquinavir [55], lopinavir/ritonavir [48], ritonavir [48], NRTIs [52, 56-58] |
Pro-inflammatory | Atazanavir/ritonavir [48] lopinavir/ritonavir[48], ritonavir [48], efavirenz [50, 51, 59], rilpivirine [50] |
Prelamin A accumulation | Protease inhibitors [44] |
Cumulatively, these data support the hypothesis that adipose tissue physiology in HIV infection varies by fat type, anatomic location, and ART use rather than lipodystrophy status or fat volume alone. Impaired adipocyte differentiation has been reported with the older PIs (ritonavir, saquinavir) and the non-nucleoside reverse transcriptase inhibitors (NNRTIs, efavirenz, rilpivirine, and nevirapine) while newer therapies (including darunavir, maraviroc and raltegravir) appear to have minimal impact on adipogenesis or mitochondria. Growing evidence suggests depletion of mitochondrial DNA (mtDNA) in the adipose tissue of ART-treated and -untreated HIV-infected persons [47, 56, 57, 60-64]. Among ART-treated persons with lipodystrophy, mtDNA is depleted in both VAT and SAT, metabolism and adipogenesis markers are decreased in SAT, and less pro-inflammatory gene expression occurs in VAT, potentially protecting it from depletion [62]. Finally, SAT pro-inflammatory cytokines increase more with efavirenz (vs lopinavir-ritonavir) in combination with tenofovir and emtricitabine [59].
HIV-1-specific factors may also contribute to adipose tissue inflammation. The HIV accessory protein viral protein R (Vpr) may induce adipose tissue dysfunction by inhibiting peroxisome proliferator-activated receptor (PPAR)-γ and activating glucocorticoid genes, leading to lipolysis, macrophage infiltration into adipose tissue, loss of white adipose depots and hepatic steatosis [65]. Additionally, adipose tissue may serve as a reservoir for HIV [66], and longer duration of both HIV and ART use were positively associated with higher levels of TNF-α, caspase-3 and TGF-β [43].
Gut mucosal destruction following HIV infection and subsequent persistent microbial translocation are well described. Drainage of microbial products to the liver may cause local inflammation, increased synthesis of triglycerides and fat droplet accumulation in hepatocytes, contributing to visceral adiposity [67, 68]. Additionally, both HIV infection and obesity are independently associated with gut microbiome alterations and increased intestinal permeability that further promote local and systemic inflammation [69-75]. Although a recent study demonstrated higher levels of monocyte activation and systemic inflammation in obese vs non-obese HIV-infected persons, additional research is needed to determine whether HIV and obesity have a synergistic effect on the burden of inflammation-related metabolic disease.
Finally, the pro-inflammatory, pro-atherogenic and pro-thrombotic renin angiotensin system may be activated in HIV infection and/or by ART [76, 77]. Renin angiotensin activity increases with VAT volume irrespective of HIV serostatus [76], but studies have been inconclusive as to whether renin angiotensin system inhibition may have beneficial effects on VAT or inflammation in HIV [78-80].
Implications of Ectopic Fat
Ectopic fat deposition is associated with inflammation and adverse metabolic impact beyond that seen with generalized obesity [81]. Associations between intra-abdominal VAT and increased metabolic disease risk (including CVD) are well described both in cross-sectional and longitudinal studies [81, 82]. In a cross-sectional study of nearly 600 HIV-infected men on stable ART, greater VAT, liver fat, and epicardial fat were independently associated with CVD after adjusting for traditional CVD risk factors [83]. HIV-infected participants in the CHARTER study with increased visceral adiposity (estimated by waist circumference ≥88 cm in women or ≥102 cm in men) had significantly worse neurocognitive function. Furthermore, an association between higher interleukin (IL)-6 levels and poorer neurocognitive function was found only among those with the largest waist circumferences, supporting a link between visceral adiposity, inflammation, and neurocognitive function in HIV-infected persons [84].
Similar adverse outcomes are found in other ectopic fat sites: in an older, HIV-uninfected population, skeletal muscle lipid content was a better predictor of insulin resistance than BMI, waist-to-hip ratio, or total body fat [85, 86]. Similarly, computed tomography (CT)-estimated paraspinal fat was a significant predictor of metabolic syndrome in women after adjustment for BMI and VAT [87]. Furthermore, skeletal muscle fat had functional consequences: greater thigh skeletal muscle fat was independently associated with lower muscle strength, slower chair rise time, and slower gait speed [88-90]; and fatty infiltration of the trunk muscles has been cross-sectionally and longitudinally associated with impaired physical function [91]. Surprisingly, HIV-infected participants in the Fat Redistribution and Metabolic Change in HIV Infection (FRAM) Study had significantly lower intermuscular adipose tissue (by magnetic resonance imaging) compared to HIV-uninfected controls, a finding that was attenuated but persisted in multivariate analyses [92]. In contrast, a greater amount of mid-thigh muscle bundle fatty infiltration (estimated by CT scan) was found among middle-aged, HIV-infected vs HIV-uninfected men after multivariable adjustment including VAT and SAT. Furthermore, the fatty infiltration increased over time among HIV-infected compared to HIV-uninfected men (2015 Annual Meeting of the Endocrine Society; San Diego, CA; Natsag et al. Abstract FRI-229). Among younger, HIV-infected persons with lipodystrophy syndrome, greater CT-estimated fatty infiltration of the psoas muscle but not BMI, SAT, lean body mass, or ART, was associated with insulin resistance [93]. These findings suggest that skeletal muscle may be a clinically relevant ectopic fat location in HIV-infected persons, impacting both metabolic and physical function outcomes. In summary, fat location may be a better marker of metabolic risk than overall adiposity, but further studies are needed to compare deposition sites among HIV-infected individuals with and without lipodystrophy.
Adipose Tissue Quality vs Quantity
Normal, healthy adipocytes are small, well differentiated and contain a modest lipid droplet. During weight gain, adipocytes become larger, less well differentiated and engorged with lipids [94, 95], making them less dense. During wasting, adipocytes become smaller and contain fewer lipids [96, 97], increasing density. Thus, changes in adipose tissue health appear to relate to changes in adipose tissue density; however, a direct comparison of (CT-quantified) adipose tissue density and histopathology has only been reported in non-human primates to date. In that study, denser VAT was associated with smaller adipocytes, lower serum leptin, and higher adiponectin levels, but not with levels of monocyte chemoattractant protein (MCP)-1, IL-6, CRP or T lymphocyte or monocyte gene expression [98].
Adipose tissue health has important implications: First, fat is an active immune and endocrine organ, and adipocyte dysfunction has been linked to pro-inflammatory cytokine expression and inflammatory diseases including insulin resistance and CVD [99]. Second, determination of adipose tissue health may aid understanding of physiologic phenomena such as metabolically healthy obesity (described below). Third, HIV-associated and ART-associated adipose tissue dysfunction are well documented and contribute to comorbid disease. Adipose tissue health in HIV remains understudied; however, ongoing ACTG and Multicenter AIDS Cohort Study investigations may help clarify how best to assess adipose tissue quality and its potential clinical implications in HIV-infected persons.
Is Fat Always Bad? Metabolically Healthy Obesity and Obesity in Older Age
Although obesity is generally associated with the development of metabolic disorders, a state of obesity without overt cardiometabolic disease, “metabolically healthy obesity”, has been described. Metabolically healthy obesity has been variably-defined in differing populations [100-102], leading to prevalence estimates ranging from 6-40% [103]. The existence of metabolically healthy obesity is controversial, but supported by the fact that metabolic dysregulation is a heterogeneous process that also occurs in the absence of obesity. Persons with metabolically healthy obesity may have less VAT and systemic inflammation and more favorable immune profiles than the metabolically unhealthy obese [104-106], further supporting a spectrum of metabolic health within obesity. In contrast, some studies have suggested increased mortality, increased CVD risk, and differences in body composition and fat distribution in HIV-uninfected persons meeting criteria for metabolically healthy obesity, suggesting that this state is not entirely benign [103, 107-112]. It is unknown whether metabolically healthy obesity exists in HIV-infected persons. Given the prevalence of metabolic syndrome in HIV (up to 45% vs 25% in the general population [113]), the metabolic effects of HIV and ART, and the persistent immune activation and inflammation associated with even virologically-suppressed HIV, it is possible that metabolically healthy obesity may not exist in HIV-infected persons, and/or that metabolic health in this population must be defined differently (2015 International Workshop on Co-morbidities and Adverse Drug Reactions in HIV; Barcelona, Spain; Lake et al. Abstract ADRLH-33).
Another paradox is that an overweight or obese body weight among older adults is associated with improved survival, and weight loss late in life can portend a poor prognosis. A strong association between mortality and weight loss has been well-described among older, HIV-uninfected adults independent of underlying disease or comorbid illnesses [114-117]. One longitudinal study observed an increased risk of death among older adults (mean age 73) who experienced any appreciable weight loss after the age of 21 and among underweight older adults in the 3rd-8th decades of life [115]. Reports among HIV-infected adults are more limited but similar in implication: in the Nutrition for Healthy Living cohort, weight loss from baseline and weight loss ≥5% over a 6-month period were significant predictors of mortality [118, 119].
Interventions to Modify Adipose Tissue in HIV
ART selection
ART initiation is associated with a gain in overall body weight that may contribute to VAT gains or lipohypertrophy [8, 23, 120-122]. Therefore, initiation of or switch to more “fat friendly” regimens is an appealing option, but should not be based upon changes in body weight only (see Implications of Ectopic Fat). For example, in ACTG study A5175, participants were randomized to efavirenz with either FTC/TDF or 3TC/ZDV. The FTC/TDF arm had significantly greater increases in overall weight as well as waist, hip, mid-arm, and mid-thigh circumferences. However, all cases of lipoatrophy occurred in the 3TC/ZDV arm, which also had greater gains in waist-hip ratio (suggestive of subtle hip lipoatrophy with relative VAT gain). Among more contemporary regimens, these differences are less apparent [10]. In ACTG study A5224s, participants randomized to ATV/r had a small but significantly greater increase in body weight than participants randomized to EFV-based therapy [123], with no significant differences seen by NRTI backbone (ABC/3TC or TDF/FTC). Although integrase inhibitors have generally been associated with smaller changes in weight and fat distribution, ACTG study A5260 observed no significant differences between raltegravir and two contemporary ritonavir-boosted PIs, darunavir or atazanavir, all with an FTC/TDF backbone (2015 Conference on Retroviruses and Opportunistic Infections; Seattle, WA; McComsey, et al. Abstract 140).
Similarly, switching ART may not reverse VAT accumulation [124-127]: HIV-infected women with central adiposity on ART switched to raltegravir from PI or NNRTI-based therapy had no significant changes in SAT or VAT area but did have significant reductions in sCD14 [126]. In the SPIRAL-LIP sub-study, participants randomized to switch from a boosted PI to raltegravir had no significant change in total adipose tissue or VAT, whereas those continuing boosted PI therapy for 48 weeks had significant increases in both [124]. In the SPIRAL parent study, switch to raltegravir was associated with significant reductions in hs-CRP, monocyte chemotactic protein (MCP)-1, IL-6, and TNF-α but not IL-10 or adiponectin, and it is not clear to what extent changes in adipose tissue explain the changes in inflammatory markers [128]. In contrast, a switch to raltegravir from stavudine therapy among lipodystrophic individuals resulted in improvements in whole body and limb fat quantity, trunk/limb fat ratio and fat mass index, with restoration of mtDNA and adiponectin levels [52].
Growth hormone (GH) axis therapies
Adiposity significantly reduces both GH secretion and pulsatility [129], thus, therapies targeting the GH axis have been of interest for obesity and lipodystrophy. Although GH has effectively decreased VAT in HIV-uninfected and -infected populations, with some studies also reporting a simultaneous reduction in CRP, reduction in VAT was at the expense of insulin resistance, multiple side effects, and loss of SAT. Thus the clinical utility of GH for obesity treatment has been limited. Of note, many initial studies administered supra-physiologic doses of continuous GH, and subsequent trials have attempted to more closely mimic physiologic levels or the pulsatile nature of GH. A subsequent, low-dose GH trial in HIV titrated doses to achieve insulin-like GH levels within the normal range, and resulted in marginally but significantly improved VAT and worsened glucose tolerance [130]. A GH releasing hormone analogue, tesamorelin, was FDA approved in 2010 after clinical trials demonstrated significant reductions in VAT with less impact on insulin resistance and no significant difference in side effects vs placebo outside of injection site reactions and edema. In contrast to the low-dose GH study where no significant changes in adiponectin or other cytokines were observed, tesamorelin use has been associated with significant improvements in adiponectin, tissue plasminogen activator (tPA) and plasminogen activator inhibitor (PAI-1) that corresponded to reductions in VAT [131, 132]. Recently, tesamorelin has also been shown to reduce VAT in obese, HIV-uninfected persons [133].
Exercise
In epidemiologic studies, regular physical activity is associated with reduced risk of inflammation-associated disease, including dementia, cancer, CVD, and insulin resistance. In contrast, prolonged physical inactivity is associated with VAT accumulation and elevation of multiple pro-inflammatory cytokine levels. Some of the benefit of exercise may be explained through alteration in systemic cytokines [134, 135], however, few recent studies have explored the impact of exercise on markers of inflammation or immune activation in HIV-infected adults, particularly in regards to body composition changes. A systemic review published in 2010 summarized the effects of exercise in HIV-infected adults, with prior studies primarily focused on wasting or lipodystrophy: aerobic exercise generally led to improvements in BMI, triceps SAT, waist circumference, and waist-to-hip ratio [136]. Since then, a factorial intervention of lifestyle modification with or without metformin among HIV-infected adults with the metabolic syndrome observed a trend towards greater VAT reductions (without SAT loss) in the metformin group, and significantly greater reductions in intra-myocellular lipid content and hs-CRP in the lifestyle modification group [137]. In a small study of 18 HIV-infected participants with lipodystrophy randomized to 16 weeks of strength or endurance training, significantly greater decreases in total and limb fat mass were seen in the strength training group, while significantly greater reductions in systemic inflammation (hs-CRP, IL-6, TNF-α, and IL-18) were observed in the endurance group, suggesting that the reduction in inflammation was not due to changes in fat mass alone [138]. Thirty-five older adults completing a 12-week intervention of endurance with or without strength training demonstrated significant decreases in IL-6, hs-CRP, d-dimer, and IL-18 (2014 Conference on Retroviruses and Opportunistic Infections; Boston, MA; Longo, et al. Abstract 763). Finally, strength training led to decreases in trunk fat and triglyceride levels, but neither strength nor cardiovascular exercise decreased lipopolysaccharide levels (a marker of microbial translocation) [139].
Other interventions
Recently, nutritional interventions, including those targeting the intestinal microbiome, have gained traction as potential therapies for adipose tissue dysfunction. In an animal model of diet-induced obesity, arginase inhibition ameliorated obesity-induced adipose tissue inflammation by reducing macrophage infiltration into adipose tissue, improving adipose tissue monocyte profiles, and decreasing pro-inflammatory cytokine expression [140]. Several studies have suggested that probiotic supplementation may help improve adipose tissue dysfunction by “resetting” the gut microbiome, which could help restore gut mucosal integrity, decrease microbial translocation, and decrease systemic inflammation. In the recent Probio-HIV study, ART-treated, HIV-infected adults treated with probiotics demonstrated reduced CD4+ T lymphocyte activation and reduced plasma levels of lipopolysaccharide binding protein and hs-CRP [141]. Similar studies have demonstrated improvements in d-dimer, IL-6 and lipopolysaccharide binding protein [142, 143]. An upcoming ACTG study will study the effect of the probiotic Visbiome on markers of microbial translocation, inflammation and immune activation in adults on suppressive ART.
Recent data suggest that trimethoprim sulfamethoxazole use at the time of ART initiation may improve some markers of microbial translocations without restoring the gut blood-barrier, a finding the authors hypothesized was mediated through modulation of the gut microbiome [144]. Finally, in an animal model of obesity, maraviroc therapy was associated with simultaneous improvements in gut Enterobacteriales, body weight gain, insulin sensitivity and liver fat [145].
Conclusions
While data on the contribution of adipose tissue dysfunction to health in treated HIV infection are emerging, many questions remain unanswered, including how adipose tissue physiology may be unique in HIV infection and/or with ART selection, the trajectory of body weight and hip/waist circumference throughout the lifetime of an HIV-infected person and/or the effects of obesity duration in HIV-infected compared to uninfected populations. Additionally, whether these associations differ by ART era and timing of ART initiation also remains unknown. Although multiple ongoing studies aim to address some of these questions, additional research is needed, particularly in regards to tissue-level physiology and the effects of newer antiretroviral agents.
Acknowledgments
This manuscript was supported through funding from the National Institutes of Health, National Institute on Aging (K23 AG050260 to KM Erlandson) and National Institutes of Allergy and Infectious Diseases (K23 AI110532 to JE Lake).
Footnotes
Conflict of Interest: Kristine M. Erlandson reports research grants to her institution through the Gilead Sciences Research Scholars Program. Jordan E Lake reports personal fees from Gilead Sciences and GlaxoSmithKline.
Compliance with Ethics Guidelines: Human and Animal Rights and Informed Consent: This article does not contain any studies with human or animal subjects performed by any of the authors.
Contributor Information
Kristine M. Erlandson, Email: kristine.erlandson@ucdenver.edu.
Jordan E. Lake, Email: jlake@mednet.ucla.edu.
References
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of childhood and adult obesity in the United States, 2011-2012. JAMA. 2014;311:806–814. doi: 10.1001/jama.2014.732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Stenholm S, Strandberg TE, Pitkala K, Sainio P, Heliovaara M, Koskinen S. Midlife obesity and risk of frailty in old age during a 22-year follow-up in men and women: the Mini-Finland Follow-up Survey. J Gerontol A Biol Sci Med Sci. 2014;69:73–78. doi: 10.1093/gerona/glt052. [DOI] [PubMed] [Google Scholar]
- 3.Strandberg TE, Sirola J, Pitkala KH, Tilvis RS, Strandberg AY, Stenholm S. Association of midlife obesity and cardiovascular risk with old age frailty: a 26-year follow-up of initially healthy men. Int J Obes (Lond) 2012;36:1153–1157. doi: 10.1038/ijo.2012.83. [DOI] [PubMed] [Google Scholar]
- 4.Crum-Cianflone N, Roediger MP, Eberly L, Headd M, Marconi V, Ganesan A, et al. Increasing rates of obesity among HIV-infected persons during the HIV epidemic. PLoS One. 2010;5:e10106. doi: 10.1371/journal.pone.0010106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Buchacz K, Baker RK, Palella FJ, Jr, Shaw L, Patel P, Lichtenstein KA, et al. Disparities in prevalence of key chronic diseases by gender and race/ethnicity among antiretroviral-treated HIV-infected adults in the US. Antivir Ther. 2013;18:65–75. doi: 10.3851/IMP2450. [DOI] [PubMed] [Google Scholar]
- 6.Wand H, Ramjee G. High prevalence of obesity among women who enrolled in HIV prevention trials in KwaZulu-Natal, South Africa: healthy diet and life style messages should be integrated into HIV prevention programs. BMC Public Health. 2013;13:159. doi: 10.1186/1471-2458-13-159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fontbonne A, Cournil A, Cames C, Mercier S, Coly AN, Lacroux A, et al. Anthropometric characteristics and cardiometabolic risk factors in a sample of urban-dwelling adults in Senegal. Diabetes Metab. 2011;37:52–58. doi: 10.1016/j.diabet.2010.07.008. [DOI] [PubMed] [Google Scholar]
- 8.Wohl DA, Brown TT. Management of morphologic changes associated with antiretroviral use in HIV-infected patients. J Acquir Immune Defic Syndr. 2008;49(2):S93–S100. doi: 10.1097/QAI.0b013e318186521a. [DOI] [PubMed] [Google Scholar]
- 9.Lakey W, Yang LY, Yancy W, Chow SC, Hicks C. Short communication: from wasting to obesity: initial antiretroviral therapy and weight gain in HIV-infected persons. AIDS Res Hum Retroviruses. 2013;29:435–440. doi: 10.1089/aid.2012.0234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Erlandson KM, Taejaroenkul S, Smeaton L, Gupta A, Singini IL, Lama JR, et al. A Randomized Comparison of Anthropomorphic Changes With Preferred and Alternative Efavirenz-Based Antiretroviral Regimens in Diverse Multinational Settings. Open Forum Infect Dis. 2015;2:ofv095. doi: 10.1093/ofid/ofv095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- •11.Ali MK, Magee MJ, Dave JA, Ofotokun I, Tungsiripat M, Jones TK, et al. HIV and Metabolic, Body, and Bone Disorders: What We Know From Low- and Middle-Income Countries. J Acquir Immune Defic Syndr. 2014;67(Suppl 1):S27–39. doi: 10.1097/QAI.0000000000000256. This supplement summarizes the existing knowledge and highlights important remaining research agenda questions specifically for low- and middle-income countries (LMICs) [DOI] [PubMed] [Google Scholar]
- 12.Tate T, Willig AL, Willig JH, Raper JL, Moneyham L, Kempf MC, et al. HIV infection and obesity: where did all the wasting go? Antivir Ther. 2012;17:1281–1289. doi: 10.3851/IMP2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Maia Leite LH, De Mattos Marinho Sampaio AB. Progression to overweight, obesity and associated factors after antiretroviral therapy initiation among Brazilian persons with HIV/AIDS. Nutr Hosp. 2010;25:635–640. [PubMed] [Google Scholar]
- •14.Yuh B, Tate J, Butt AA, Crothers K, Freiberg M, Leaf D, et al. Weight change after antiretroviral therapy and mortality. Clin Infect Dis. 2015;60:1852–1859. doi: 10.1093/cid/civ192. From the Veterans Aging Cohort Study, this is one of the first publications in the current ART era to show the potential consequences of weight gain after ART initiation. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kim DJ, Westfall AO, Chamot E, Willig AL, Mugavero MJ, Ritchie C, et al. Multimorbidity patterns in HIV-infected patients: the role of obesity in chronic disease clustering. J Acquir Immune Defic Syndr. 2012;61:600–605. doi: 10.1097/QAI.0b013e31827303d5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sudfeld CR, Isanaka S, Mugusi FM, Aboud S, Wang M, Chalamilla GE, et al. Weight change at 1 mo of antiretroviral therapy and its association with subsequent mortality, morbidity, and CD4 T cell reconstitution in a Tanzanian HIV-infected adult cohort. Am J Clin Nutr. 2013;97:1278–1287. doi: 10.3945/ajcn.112.053728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Menucci MB, Burman KD. Endocrine Changes in Obesity. In: De Groot LJ, Beck-Peccoz P, Chrousos G, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000. [Google Scholar]
- 18.Koster A, Ding J, Stenholm S, Caserotti P, Houston DK, Nicklas BJ, et al. Does the amount of fat mass predict age-related loss of lean mass, muscle strength, and muscle quality in older adults? J Gerontol A Biol Sci Med Sci. 2011;66:888–895. doi: 10.1093/gerona/glr070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bauer LO, Wu Z, Wolfson LI. An obese body mass increases the adverse effects of HIV/AIDS on balance and gait. Phys Ther. 2011;91:1063–1071. doi: 10.2522/ptj.20100292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shah K, Hilton TN, Myers L, Pinto JF, Luque AE, Hall WJ. A new frailty syndrome: central obesity and frailty in older adults with the human immunodeficiency virus. J Am Geriatr Soc. 2012;60:545–549. doi: 10.1111/j.1532-5415.2011.03819.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Erlandson KM, Allshouse AA, Jankowski CM, Duong S, Mawhinney S, Kohrt WM, et al. Comparison of functional status instruments in HIV-infected adults on effective antiretroviral therapy. HIV clinical trials. 2012;13:324–334. doi: 10.1310/hct1306-324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Freitas P, Carvalho D, Santos AC, Madureira AJ, Martinez E, Pereira J, et al. Carotid intima media thickness is associated with body fat abnormalities in HIV-infected patients. BMC Infect Dis. 2014;14:348. doi: 10.1186/1471-2334-14-348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Haubrich RH, Riddler SA, DiRienzo AG, Komarow L, Powderly WG, Klingman K, et al. Metabolic outcomes in a randomized trial of nucleoside, nonnucleoside and protease inhibitor-sparing regimens for initial HIV treatment. AIDS. 2009;23:1109–1118. doi: 10.1097/QAD.0b013e32832b4377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Dube MP, Komarow L, Mulligan K, Grinspoon SK, Parker RA, Robbins GK, et al. Long-term body fat outcomes in antiretroviral-naive participants randomized to nelfinavir or efavirenz or both plus dual nucleosides. Dual X-ray absorptiometry results from A5005s, a substudy of Adult Clinical Trials Group 384. J Acquir Immune Defic Syndr. 2007;45:508–514. doi: 10.1097/QAI.0b013e3181142d26. [DOI] [PubMed] [Google Scholar]
- 25.Mutimura E, Stewart A, Rheeder P, Crowther NJ. Metabolic function and the prevalence of lipodystrophy in a population of HIV-infected African subjects receiving highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2007;46:451–455. doi: 10.1097/qai.0b013e318158c0a6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shah RV, Murthy VL, Abbasi SA, Blankstein R, Kwong RY, Goldfine AB, et al. Visceral adiposity and the risk of metabolic syndrome across body mass index: the MESA Study. JACC Cardiovasc Imaging. 2014;7:1221–1235. doi: 10.1016/j.jcmg.2014.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Scherzer R, Heymsfield SB, Lee D, Powderly WG, Tien PC, Bacchetti P, et al. Decreased limb muscle and increased central adiposity are associated with 5-year all-cause mortality in HIV infection. AIDS. 2011;25:1405–1414. doi: 10.1097/QAD.0b013e32834884e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Guaraldi G, Murri R, Orlando G, Squillace N, Stentarelli C, Zona S, et al. Lipodystrophy and quality of life of HIV-infected persons. AIDS Rev. 2008;10:152–161. [PubMed] [Google Scholar]
- 29.Reynolds NR, Neidig JL, Wu AW, Gifford AL, Holmes WC. Balancing disfigurement and fear of disease progression: Patient perceptions of HIV body fat redistribution. AIDS Care. 2006;18:663–673. doi: 10.1080/09540120500287051. [DOI] [PubMed] [Google Scholar]
- 30.Guaraldi G, Murri R, Orlando G, Giovanardi C, Squillace N, Vandelli M, et al. Severity of lipodystrophy is associated with decreased health-related quality of life. AIDS Patient Care STDS. 2008;22:577–585. doi: 10.1089/apc.2007.0173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Burgoyne R, Collins E, Wagner C, Abbey S, Halman M, Nur M, et al. The relationship between lipodystrophy-associated body changes and measures of quality of life and mental health for HIV-positive adults. Qual Life Res. 2005;14:981–990. doi: 10.1007/s11136-004-2580-2. [DOI] [PubMed] [Google Scholar]
- 32.Lenert LA, Feddersen M, Sturley A, Lee D. Adverse effects of medications and tradeoffs between length of life and quality of life in human immunodeficiency virus infection. Am J Med. 2002;113:229–232. doi: 10.1016/s0002-9343(02)01156-7. [DOI] [PubMed] [Google Scholar]
- 33.Poudel-Tandukar K, Bertone-Johnson ER, Palmer PH, Poudel KC. C-reactive protein and depression in persons with Human Immunodeficiency Virus infection: The Positive Living with HIV (POLH) Study. Brain Behav Immun. 2014;42:89–95. doi: 10.1016/j.bbi.2014.06.004. [DOI] [PubMed] [Google Scholar]
- 34.Koethe JR, Dee K, Bian A, Shintani A, Turner M, Bebawy S, et al. Circulating interleukin-6, soluble CD14, and other inflammation biomarker levels differ between obese and nonobese HIV-infected adults on antiretroviral therapy. AIDS Res Hum Retroviruses. 2013;29:1019–1025. doi: 10.1089/aid.2013.0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Betene ADC, De Wit S, Neuhaus J, Palfreeman A, Pepe R, Pankow JS, et al. Interleukin-6, High Sensitivity C-Reactive Protein, and the Development of Type 2 Diabetes Among HIV-Positive Patients Taking Antiretroviral Therapy. J Acquir Immune Defic Syndr. 2014;67:538–546. doi: 10.1097/QAI.0000000000000354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bourlier V, Sengenes C, Zakaroff-Girard A, Decaunes P, Wdziekonski B, Galitzky J, et al. TGFbeta family members are key mediators in the induction of myofibroblast phenotype of human adipose tissue progenitor cells by macrophages. PLoS One. 2012;7:e31274. doi: 10.1371/journal.pone.0031274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Haase J, Weyer U, Immig K, Kloting N, Bluher M, Eilers J, et al. Local proliferation of macrophages in adipose tissue during obesity-induced inflammation. Diabetologia. 2014;57:562–571. doi: 10.1007/s00125-013-3139-y. [DOI] [PubMed] [Google Scholar]
- 38.Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17:179–188. doi: 10.1038/nm.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ••39.Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161:146–160. doi: 10.1016/j.cell.2015.02.022. This is an excellent review on the interaction between the immune system and adiposity. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hong KM, Burdick MD, Phillips RJ, Heber D, Strieter RM. Characterization of human fibrocytes as circulating adipocyte progenitors and the formation of human adipose tissue in SCID mice. FASEB J. 2005;19:2029–2031. doi: 10.1096/fj.05-4295fje. [DOI] [PubMed] [Google Scholar]
- 41.Divoux A, Tordjman J, Lacasa D, Veyrie N, Hugol D, Aissat A, et al. Fibrosis in human adipose tissue: composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes. 2010;59:2817–2825. doi: 10.2337/db10-0585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Giralt M, Domingo P, Villarroya F. Adipose tissue biology and HIV-infection. Best Pract Res Clin Endocrinol Metab. 2011;25:487–499. doi: 10.1016/j.beem.2010.12.001. [DOI] [PubMed] [Google Scholar]
- 43.de Souza Dantes, Oliveira SdSA TL, da Silva Barbosa L, Souza Lisboa PG, Tavares Dutra CD, Margalho Sousa L, Simoes Quaresma JA, Feio Libonati RM. Immunohistochemical analysis of the expression of TNF-alpha, TGF-beta, and caspase-3 in subcutaneous tissue of patients with HIV lipodystrophy syndrome. Microb Pathog. 2014:41–47. doi: 10.1016/j.micpath.2014.02.004. [DOI] [PubMed] [Google Scholar]
- 44.Bereziat V, Cervera P, Le Dour C, Verpont MC, Dumont S, Vantyghem MC, et al. LMNA mutations induce a non-inflammatory fibrosis and a brown fat-like dystrophy of enlarged cervical adipose tissue. Am J Pathol. 2011;179:2443–2453. doi: 10.1016/j.ajpath.2011.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Torriani M, Fitch K, Stavrou E, Bredella MA, Lim R, Sass CA, et al. Deiodinase 2 expression is increased in dorsocervical fat of patients with HIV-associated lipohypertrophy syndrome. J Clin Endocrinol Metab. 2012;97:E602–607. doi: 10.1210/jc.2011-2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kosmiski LA, Sage-El A, Kealey EH, Bessesen DH. Brown fat activity is not apparent in subjects with HIV lipodystrophy and increased resting energy expenditure. Obesity (Silver Spring) 2011;19:2096–2098. doi: 10.1038/oby.2011.231. [DOI] [PubMed] [Google Scholar]
- 47.Sevastianova K, Sutinen J, Greco D, Sievers M, Salmenkivi K, Perttila J, et al. Comparison of dorsocervical with abdominal subcutaneous adipose tissue in patients with and without antiretroviral therapy-associated lipodystrophy. Diabetes. 2011;60:1894–1900. doi: 10.2337/db11-0075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Capel E, Auclair M, Caron-Debarle M, Capeau J. Effects of ritonavir-boosted darunavir, atazanavir and lopinavir on adipose functions and insulin sensitivity in murine and human adipocytes. Antivir Ther. 2012;17:549–556. doi: 10.3851/IMP1988. [DOI] [PubMed] [Google Scholar]
- 49.Kitazawa T, Yoshino Y, Suzuki S, Koga I, Ota Y. Lopinavir inhibits insulin signaling by promoting protein tyrosine phosphatase 1B expression. Exp Ther Med. 2014;8:851–855. doi: 10.3892/etm.2014.1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Diaz-Delfin J, Domingo P, Mateo MG, Gutierrez Mdel M, Domingo JC, Giralt M, et al. Effects of rilpivirine on human adipocyte differentiation, gene expression, and release of adipokines and cytokines. Antimicrob Agents Chemother. 2012;56:3369–3375. doi: 10.1128/AAC.00104-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Diaz-Delfin J, del Mar Gutierrez M, Gallego-Escuredo JM, Domingo JC, Gracia Mateo M, Villarroya F, et al. Effects of nevirapine and efavirenz on human adipocyte differentiation, gene expression, and release of adipokines and cytokines. Antiviral Res. 2011;91:112–119. doi: 10.1016/j.antiviral.2011.04.018. [DOI] [PubMed] [Google Scholar]
- 52.Domingo P, Gutierrez Mdel M, Gallego-Escuredo JM, Torres F, Mateo GM, Villarroya J, et al. Effects of switching from stavudine to raltegravir on subcutaneous adipose tissue in HIV-infected patients with HIV/HAART-associated lipodystrophy syndrome (HALS). A clinical and molecular study. PLoS One. 2014;9:e89088. doi: 10.1371/journal.pone.0089088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gibellini L, De Biasi S, Pinti M, Nasi M, Riccio M, Carnevale G, et al. The protease inhibitor atazanavir triggers autophagy and mitophagy in human preadipocytes. AIDS. 2012;26:2017–2026. doi: 10.1097/QAD.0b013e328359b8be. [DOI] [PubMed] [Google Scholar]
- 54.Minami R, Yamamoto M, Takahama S, Ando H, Miyamura T, Suematsu E. Comparison of the influence of four classes of HIV antiretrovirals on adipogenic differentiation: the minimal effect of raltegravir and atazanavir. J Infect Chemother. 2011;17:183–188. doi: 10.1007/s10156-010-0101-5. [DOI] [PubMed] [Google Scholar]
- 55.Bociaga-Jasik M, Polus A, Goralska J, Czech U, Gruca A, Sliwa A, et al. Metabolic effects of the HIV protease inhibitor--saquinavir in differentiating human preadipocytes. Pharmacol Rep. 2013;65:937–950. doi: 10.1016/s1734-1140(13)71075-2. [DOI] [PubMed] [Google Scholar]
- 56.Feeney ER, van Vonderen MG, Wit F, Danner SA, van Agtmael MA, Villarroya F, et al. Zidovudine/lamivudine but not nevirapine in combination with lopinavir/ritonavir decreases subcutaneous adipose tissue mitochondrial DNA. AIDS. 2012;26:2165–2174. doi: 10.1097/QAD.0b013e328358b279. [DOI] [PubMed] [Google Scholar]
- 57.McComsey GA, Daar ES, O'Riordan M, Collier AC, Kosmiski L, Santana JL, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Menezes CN, Duarte R, Dickens C, Dix-Peek T, Van Amsterdam D, John MA, et al. The early effects of stavudine compared with tenofovir on adipocyte gene expression, mitochondrial DNA copy number and metabolic parameters in South African HIV-infected patients: a randomized trial. HIV Med. 2013;14:217–225. doi: 10.1111/j.1468-1293.2012.01054.x. [DOI] [PubMed] [Google Scholar]
- 59.Domingo P, Gutierrez Mdel M, Gallego-Escuredo JM, Torres F, Mateo MG, Villarroya J, et al. A 48-week study of fat molecular alterations in HIV naive patients starting tenofovir/emtricitabine with lopinavir/ritonavir or efavirenz. J Acquir Immune Defic Syndr. 2014;66:457–465. doi: 10.1097/QAI.0000000000000205. [DOI] [PubMed] [Google Scholar]
- 60.Garrabou G, Lopez S, Moren C, Martinez E, Fontdevila J, Cardellach F, et al. Mitochondrial damage in adipose tissue of untreated HIV-infected patients. AIDS. 2011;25:165–170. doi: 10.1097/QAD.0b013e3283423219. [DOI] [PubMed] [Google Scholar]
- 61.Vidal F, Domingo P, Villarroya F, Giralt M, Lopez-Dupla M, Gutierrez M, et al. Adipogenic/lipid, inflammatory, and mitochondrial parameters in subcutaneous adipose tissue of untreated HIV-1-infected long-term nonprogressors: significant alterations despite low viral burden. J Acquir Immune Defic Syndr. 2012;61:131–137. doi: 10.1097/QAI.0b013e31825c3a68. [DOI] [PubMed] [Google Scholar]
- 62.Gallego-Escuredo JM, Villarroya J, Domingo P, Targarona EM, Alegre M, Domingo JC, et al. Differentially altered molecular signature of visceral adipose tissue in HIV-1-associated lipodystrophy. J Acquir Immune Defic Syndr. 2013;64:142–148. doi: 10.1097/QAI.0b013e31829bdb67. [DOI] [PubMed] [Google Scholar]
- 63.McGee KC, Shahmanesh M, Boothby M, Nightingale P, Gathercole LL, Tripathi G, et al. Evidence for a shift to anaerobic metabolism in adipose tissue in efavirenz-containing regimens for HIV with different nucleoside backbones. Antivir Ther. 2012;17:495–507. doi: 10.3851/IMP2017. [DOI] [PubMed] [Google Scholar]
- 64.Morse CG, Voss JG, Rakocevic G, McLaughlin M, Vinton CL, Huber 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]
- 65.Agarwal N, Balasubramanyam A. Viral mechanisms of adipose dysfunction: lessons from HIV-1 Vpr. Adipocyte. 2015;4:55–59. doi: 10.4161/adip.29852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- •66.Couturier J, Suliburk JW, Brown JM, Luke DJ, Agarwal N, Yu X, et al. Human adipose tissue as a reservoir for memory CD4+ T cells and HIV. AIDS. 2015;29:667–674. doi: 10.1097/QAD.0000000000000599. This article highlights adipose tissue as an immune organ and potential reservoir for HIV, an important consideration for the HIV cure agenda. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.van Wijk JP, Cabezas MC. Hypertriglyceridemia, Metabolic Syndrome, and Cardiovascular Disease in HIV-Infected Patients: Effects of Antiretroviral Therapy and Adipose Tissue Distribution. Int J Vasc Med. 2012;2012:201027. doi: 10.1155/2012/201027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Balagopal A, Philp FH, Astemborski J, Block TM, Mehta A, Long R, et al. Human immunodeficiency virus-related microbial translocation and progression of hepatitis C. Gastroenterology. 2008;135:226–233. doi: 10.1053/j.gastro.2008.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dillon SM, Lee EJ, Kotter CV, Austin GL, Gianella S, Siewe B, et al. Gut dendritic cell activation links an altered colonic microbiome to mucosal and systemic T-cell activation in untreated HIV-1 infection. Mucosal Immunol. 2015 doi: 10.1038/mi.2015.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Vesterbacka J, Barqasho B, Haggblom A, Nowak P. Effects of Co-Trimoxazole on Microbial Translocation in HIV-1-Infected Patients Initiating Antiretroviral Therapy. AIDS Res Hum Retroviruses. 2015 doi: 10.1089/AID.2014.0366. [DOI] [PubMed] [Google Scholar]
- 71.Vujkovic-Cvijin I, Dunham RM, Iwai S, Maher MC, Albright RG, Broadhurst MJ, et al. Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Sci Transl Med. 2013;5:193ra191. doi: 10.1126/scitranslmed.3006438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lozupone CA, Rhodes ME, Neff CP, Fontenot AP, Campbell TB, Palmer BE. HIV-induced alteration in gut microbiota: driving factors, consequences, and effects of antiretroviral therapy. Gut Microbes. 2014;5:562–570. doi: 10.4161/gmic.32132. [DOI] [PubMed] [Google Scholar]
- 73.Perez-Santiago J, Gianella S, Massanella M, Spina CA, Karris MY, Var SR, et al. Gut Lactobacillales are associated with higher CD4 and less microbial translocation during HIV infection. AIDS. 2013;27:1921–1931. doi: 10.1097/qad.0b013e3283611816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Murphy EA, Velazquez KT, Herbert KM. Influence of high-fat diet on gut microbiota: a driving force for chronic disease risk. Curr Opin Clin Nutr Metab Care. 2015 doi: 10.1097/MCO.0000000000000209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kirpich IA, Marsano LS, McClain CJ. Gut-liver axis, nutrition, and non-alcoholic fatty liver disease. Clin Biochem. 2015 doi: 10.1016/j.clinbiochem.2015.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Srinivasa S, Fitch KV, Wong K, Torriani M, Mayhew C, Stanley T, et al. RAAS Activation Is Associated With Visceral Adiposity and Insulin Resistance Among HIV-infected Patients. J Clin Endocrinol Metab. 2015:jc20151461. doi: 10.1210/jc.2015-1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Boccara F, Auclair M, Cohen A, Lefevre C, Prot M, Bastard JP, et al. HIV protease inhibitors activate the adipocyte renin angiotensin system. Antivir Ther. 2010;15:363–375. doi: 10.3851/IMP1533. [DOI] [PubMed] [Google Scholar]
- 78.Lake JE, Tseng CH, Currier JS. A pilot study of telmisartan for visceral adiposity in HIV infection: the metabolic abnormalities, telmisartan, and HIV infection (MATH) trial. PLoS One. 2013;8:e58135. doi: 10.1371/journal.pone.0058135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ucciferri C, Falasca K, Mancino P, Di Iorio A, Vecchiet J. Microalbuminuria and hypertension in HIV-infected patients: a preliminary study of telmisartan. Eur Rev Med Pharmacol Sci. 2012;16:491–498. [PubMed] [Google Scholar]
- 80.Vecchiet J, Ucciferri C, Falasca K, Mancino P, Di Iorio A, De Caterina R. Antihypertensive and metabolic effects of telmisartan in hypertensive HIV-positive patients. Antivir Ther. 2011;16:639–645. doi: 10.3851/IMP1809. [DOI] [PubMed] [Google Scholar]
- •81.Lim S, Meigs JB. Links between ectopic fat and vascular disease in humans. Arterioscler Thromb Vasc Biol. 2014;34:1820–1826. doi: 10.1161/ATVBAHA.114.303035. This article draws attention to the importance of fat distribution to metabolic health, focusing on the important link between fat maldistribution and cardiovascular disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Alexopoulos N, Katritsis D, Raggi P. Visceral adipose tissue as a source of inflammation and promoter of atherosclerosis. Atherosclerosis. 2014;233:104–112. doi: 10.1016/j.atherosclerosis.2013.12.023. [DOI] [PubMed] [Google Scholar]
- 83.Orlando G, Guaraldi G, Zona S, Carli F, Bagni P, Menozzi M, et al. Ectopic fat is linked to prior cardiovascular events in men with HIV. J Acquir Immune Defic Syndr. 2012;59:494–497. doi: 10.1097/QAI.0b013e31824c8397. [DOI] [PubMed] [Google Scholar]
- 84.Sattler FR, He J, Letendre S, Wilson C, Sanders C, Heaton R, et al. Abdominal obesity contributes to neurocognitive impairment in HIV-infected patients with increased inflammation and immune activation. J Acquir Immune Defic Syndr. 2015;68:281–288. doi: 10.1097/QAI.0000000000000458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Sinha R, Dufour S, Petersen KF, LeBon V, Enoksson S, Ma YZ, et al. Assessment of skeletal muscle triglyceride content by (1)H nuclear magnetic resonance spectroscopy in lean and obese adolescents: relationships to insulin sensitivity, total body fat, and central adiposity. Diabetes. 2002;51:1022–1027. doi: 10.2337/diabetes.51.4.1022. [DOI] [PubMed] [Google Scholar]
- 86.Krssak M, Falk Petersen K, Dresner A, DiPietro L, Vogel SM, Rothman DL, et al. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia. 1999;42:113–116. doi: 10.1007/s001250051123. [DOI] [PubMed] [Google Scholar]
- 87.Therkelsen KE, Pedley A, Speliotes EK, Massaro JM, Murabito J, Hoffmann U, et al. Intramuscular fat and associations with metabolic risk factors in the Framingham Heart Study. Arterioscler Thromb Vasc Biol. 2013;33:863–870. doi: 10.1161/ATVBAHA.112.301009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lang T, Cauley JA, Tylavsky F, Bauer D, Cummings S, Harris TB, et al. Computed tomographic measurements of thigh muscle cross-sectional area and attenuation coefficient predict hip fracture: the health, aging, and body composition study. J Bone Miner Res. 2010;25:513–519. doi: 10.1359/jbmr.090807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Goodpaster BH, Carlson CL, Visser M, Kelley DE, Scherzinger A, Harris TB, et al. Attenuation of skeletal muscle and strength in the elderly: The Health ABC Study. J Appl Physiol (1985) 2001;90:2157–2165. doi: 10.1152/jappl.2001.90.6.2157. [DOI] [PubMed] [Google Scholar]
- 90.Visser M, Goodpaster BH, Kritchevsky SB, Newman AB, Nevitt M, Rubin SM, et al. Muscle mass, muscle strength, and muscle fat infiltration as predictors of incident mobility limitations in well-functioning older persons. J Gerontol A Biol Sci Med Sci. 2005;60:324–333. doi: 10.1093/gerona/60.3.324. [DOI] [PubMed] [Google Scholar]
- 91.Hicks GE, Simonsick EM, Harris TB, Newman AB, Weiner DK, Nevitt MA, et al. Trunk muscle composition as a predictor of reduced functional capacity in the health, aging and body composition study: the moderating role of back pain. J Gerontol A Biol Sci Med Sci. 2005;60:1420–1424. doi: 10.1093/gerona/60.11.1420. [DOI] [PubMed] [Google Scholar]
- 92.Scherzer R, Shen W, Heymsfield SB, Lewis CE, Kotler DP, Punyanitya M, et al. Intermuscular adipose tissue and metabolic associations in HIV infection. Obesity (Silver Spring) 2011;19:283–291. doi: 10.1038/oby.2010.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Torriani M, Hadigan C, Jensen ME, Grinspoon S. Psoas muscle attenuation measurement with computed tomography indicates intramuscular fat accumulation in patients with the HIV-lipodystrophy syndrome. J Appl Physiol (1985) 2003;95:1005–1010. doi: 10.1152/japplphysiol.00366.2003. [DOI] [PubMed] [Google Scholar]
- 94.Alligier M, Meugnier E, Debard C, Lambert-Porcheron S, Chanseaume E, Sothier M, et al. Subcutaneous adipose tissue remodeling during the initial phase of weight gain induced by overfeeding in humans. J Clin Endocrinol Metab. 2012;97:E183–192. doi: 10.1210/jc.2011-2314. [DOI] [PubMed] [Google Scholar]
- 95.Pasarica M, Gowronska-Kozak B, Burk D, Remedios I, Hymel D, Gimble J, et al. Adipose tissue collagen VI in obesity. J Clin Endocrinol Metab. 2009;94:5155–5162. doi: 10.1210/jc.2009-0947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mracek T, Stephens NA, Gao D, Bao Y, Ross JA, Ryden M, et al. Enhanced ZAG production by subcutaneous adipose tissue is linked to weight loss in gastrointestinal cancer patients. Br J Cancer. 2011;104:441–447. doi: 10.1038/sj.bjc.6606083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Dahlman I, Mejhert N, Linder K, Agustsson T, Mutch DM, Kulyte A, et al. Adipose tissue pathways involved in weight loss of cancer cachexia. Br J Cancer. 2010;102:1541–1548. doi: 10.1038/sj.bjc.6605665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- •98.Murphy RA, Register TC, Shively CA, Carr JJ, Ge Y, Heilbrun ME, et al. Adipose tissue density, a novel biomarker predicting mortality risk in older adults. J Gerontol A Biol Sci Med Sci. 2014;69:109–117. doi: 10.1093/gerona/glt070. This article addresses adipose tissue density as a potential marker of fat health, an important but currently under-explored area in both HIV infection and the general population. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89:2548–2556. doi: 10.1210/jc.2004-0395. [DOI] [PubMed] [Google Scholar]
- 100.Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, et al. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation. 2005;112:2735–2752. doi: 10.1161/CIRCULATIONAHA.105.169404. [DOI] [PubMed] [Google Scholar]
- 101.Wildman RP, Muntner P, Reynolds K, McGinn AP, Rajpathak S, Wylie-Rosett J, et al. The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes among the US population (NHANES 1999-2004) Arch Intern Med. 2008;168:1617–1624. doi: 10.1001/archinte.168.15.1617. [DOI] [PubMed] [Google Scholar]
- 102.Sung KC, Cha SC, Sung JW, So MS, Byrne CD. Metabolically healthy obese subjects are at risk of fatty liver but not of pre-clinical atherosclerosis. Nutr Metab Cardiovasc Dis. 2014;24:256–262. doi: 10.1016/j.numecd.2013.07.005. [DOI] [PubMed] [Google Scholar]
- 103.Phillips CM. Metabolically healthy obesity: definitions, determinants and clinical implications. Rev Endocr Metab Disord. 2013;14:219–227. doi: 10.1007/s11154-013-9252-x. [DOI] [PubMed] [Google Scholar]
- 104.Lynch LA, O'Connell JM, Kwasnik AK, Cawood TJ, O'Farrelly C, O'Shea DB. Are natural killer cells protecting the metabolically healthy obese patient? Obesity (Silver Spring) 2009;17:601–605. doi: 10.1038/oby.2008.565. [DOI] [PubMed] [Google Scholar]
- 105.Phillips CM, Perry IJ. Does inflammation determine metabolic health status in obese and nonobese adults? J Clin Endocrinol Metab. 2013;98:E1610–1619. doi: 10.1210/jc.2013-2038. [DOI] [PubMed] [Google Scholar]
- 106.Primeau V, Coderre L, Karelis AD, Brochu M, Lavoie ME, Messier V, et al. Characterizing the profile of obese patients who are metabolically healthy. Int J Obes (Lond) 2011;35:971–981. doi: 10.1038/ijo.2010.216. [DOI] [PubMed] [Google Scholar]
- 107.Roberson LL, Aneni EC, Maziak W, Agatston A, Feldman T, Rouseff M, et al. Beyond BMI: The “Metabolically healthy obese” phenotype & its association with clinical/subclinical cardiovascular disease and all-cause mortality -- a systematic review. BMC Public Health. 2014;14:14. doi: 10.1186/1471-2458-14-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Camhi SM, Katzmarzyk PT. Differences in body composition between metabolically healthy obese and metabolically abnormal obese adults. Int J Obes (Lond) 2013 doi: 10.1038/ijo.2013.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hinnouho GM, Czernichow S, Dugravot A, Batty GD, Kivimaki M, Singh-Manoux A. Metabolically healthy obesity and risk of mortality: does the definition of metabolic health matter? Diabetes Care. 2013;36:2294–2300. doi: 10.2337/dc12-1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kim TN, Park MS, Yang SJ, Yoo HJ, Kang HJ, Song W, et al. Body size phenotypes and low muscle mass: the Korean sarcopenic obesity study (KSOS) J Clin Endocrinol Metab. 2013;98:811–817. doi: 10.1210/jc.2012-3292. [DOI] [PubMed] [Google Scholar]
- 111.van der AD, Nooyens AC, van Duijnhoven FJ, Verschuren MM, Boer JM. All-cause mortality risk of metabolically healthy abdominal obese individuals: the EPIC-MORGEN study. Obesity (Silver Spring) 2014;22:557–564. doi: 10.1002/oby.20480. [DOI] [PubMed] [Google Scholar]
- 112.Kramer CK, Zinman B, Retnakaran R. Are metabolically healthy overweight and obesity benign conditions?: A systematic review and meta-analysis. Ann Intern Med. 2013;159:758–769. doi: 10.7326/0003-4819-159-11-201312030-00008. [DOI] [PubMed] [Google Scholar]
- 113.Paula AA, Falcao MC, Pacheco AG. Metabolic syndrome in HIV-infected individuals: underlying mechanisms and epidemiological aspects. AIDS Res Ther. 2013;10:32. doi: 10.1186/1742-6405-10-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Dahl AK, Fauth EB, Ernsth-Bravell M, Hassing LB, Ram N, Gerstof D. Body mass index, change in body mass index, and survival in old and very old persons. J Am Geriatr Soc. 2013;61:512–518. doi: 10.1111/jgs.12158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Corrada MM, Kawas CH, Mozaffar F, Paganini-Hill A. Association of body mass index and weight change with all-cause mortality in the elderly. Am J Epidemiol. 2006;163:938–949. doi: 10.1093/aje/kwj114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Lee CG, Boyko EJ, Nielson CM, Stefanick ML, Bauer DC, Hoffman AR, et al. Mortality risk in older men associated with changes in weight, lean mass, and fat mass. J Am Geriatr Soc. 2011;59:233–240. doi: 10.1111/j.1532-5415.2010.03245.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Harrington M, Gibson S, Cottrell RC. A review and meta-analysis of the effect of weight loss on all-cause mortality risk. Nutr Res Rev. 2009;22:93–108. doi: 10.1017/S0954422409990035. [DOI] [PubMed] [Google Scholar]
- 118.Tang AM, Forrester J, Spiegelman D, Knox TA, Tchetgen E, Gorbach SL. Weight loss and survival in HIV-positive patients in the era of highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2002;31:230–236. doi: 10.1097/00126334-200210010-00014. [DOI] [PubMed] [Google Scholar]
- 119.Mangili A, Murman DH, Zampini AM, Wanke CA. Nutrition and HIV infection: review of weight loss and wasting in the era of highly active antiretroviral therapy from the nutrition for healthy living cohort. Clin Infect Dis. 2006;42:836–842. doi: 10.1086/500398. [DOI] [PubMed] [Google Scholar]
- 120.McComsey G, Rightmire A, Wirtz V, Yang R, Mathew M, McGrath D. Changes in body composition with ritonavir-boosted and unboosted atazanavir treatment in combination with Lamivudine and Stavudine: a 96-week randomized, controlled study. Clin Infect Dis. 2009;48:1323–1326. doi: 10.1086/597776. [DOI] [PubMed] [Google Scholar]
- 121.Brown TT, Chu H, Wang Z, Palella FJ, Kingsley L, Witt MD, et al. Longitudinal increases in waist circumference are associated with HIV-serostatus, independent of antiretroviral therapy. AIDS. 2007;21:1731–1738. doi: 10.1097/QAD.0b013e328270356a. [DOI] [PubMed] [Google Scholar]
- 122.McComsey GA, Kitch D, Sax PE, Tebas P, Tierney C, Jahed NC, et al. Peripheral and central fat changes in subjects randomized to abacavir-lamivudine or tenofovir-emtricitabine with atazanavir-ritonavir or efavirenz: ACTG Study A5224s. Clin Infect Dis. 2011;53:185–196. doi: 10.1093/cid/cir324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Erlandson KM, Kitch D, Tierney C, Sax PE, Daar ES, Tebas P, et al. Weight and lean body mass change with antiretroviral initiation and impact on bone mineral density. AIDS. 2013;27:2069–2079. doi: 10.1097/QAD.0b013e328361d25d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Curran A, Martinez E, Saumoy M, del Rio L, Crespo M, Larrousse M, et al. Body composition changes after switching from protease inhibitors to raltegravir: SPIRAL-LIP substudy. AIDS. 2012;26:475–481. doi: 10.1097/QAD.0b013e32834f3507. [DOI] [PubMed] [Google Scholar]
- 125.Lake JE, McComsey GA, Hulgan TM, Wanke CA, Mangili A, Walmsley SL, et al. A randomized trial of Raltegravir replacement for protease inhibitor or non-nucleoside reverse transcriptase inhibitor in HIV-infected women with lipohypertrophy. AIDS patient care and STDs. 2012;26:532–540. doi: 10.1089/apc.2012.0135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lake JE, McComsey GA, Hulgan T, Wanke CA, Mangili A, Walmsley SL, et al. Switch to raltegravir decreases soluble CD14 in virologically suppressed overweight women: the Women, Integrase and Fat Accumulation Trial. HIV Med. 2014;15:431–441. doi: 10.1111/hiv.12128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Lake JE, McComsey GA, Hulgan T, Wanke CA, Mangili A, Walmsley SL, et al. Switch to Raltegravir From Protease Inhibitor or Nonnucleoside Reverse-Transcriptase Inhibitor Does not Reduce Visceral Fat In Human Immunodeficiency Virus-Infected Women With Central Adiposity. Open Forum Infect Dis. 2015;2:ofv059. doi: 10.1093/ofid/ofv059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Martinez E, D'Albuquerque PM, Llibre JM, Gutierrez F, Podzamczer D, Antela A, et al. Changes in cardiovascular biomarkers in HIV-infected patients switching from ritonavir-boosted protease inhibitors to raltegravir. AIDS. 2012;26:2315–2326. doi: 10.1097/QAD.0b013e328359f29c. [DOI] [PubMed] [Google Scholar]
- 129.Veldhuis JD, Anderson SM, Shah N, Bray M, Vick T, Gentili A, et al. Neurophysiological regulation and target-tissue impact of the pulsatile mode of growth hormone secretion in the human. Growth Horm IGF Res. 2001;11(A):S25–37. doi: 10.1016/s1096-6374(01)80005-8. [DOI] [PubMed] [Google Scholar]
- 130.Lo J, You SM, Canavan B, Liebau J, Beltrani G, Koutkia P, et al. Low-dose physiological growth hormone in patients with HIV and abdominal fat accumulation: a randomized controlled trial. JAMA : the journal of the American Medical Association. 2008;300:509–519. doi: 10.1001/jama.300.5.509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Stanley TL, Falutz J, Mamputu JC, Soulban G, Potvin D, Grinspoon SK. Effects of tesamorelin on inflammatory markers in HIV patients with excess abdominal fat: relationship with visceral adipose reduction. AIDS. 2011;25:1281–1288. doi: 10.1097/QAD.0b013e328347f3f1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Falutz J, Allas S, Kotler D, Thompson M, Koutkia P, Albu J, et al. A placebo-controlled, dose-ranging study of a growth hormone releasing factor in HIV-infected patients with abdominal fat accumulation. AIDS. 2005;19:1279–1287. doi: 10.1097/01.aids.0000180099.35146.30. [DOI] [PubMed] [Google Scholar]
- 133.Stanley TL, Grinspoon SK. Effects of growth hormone-releasing hormone on visceral fat, metabolic, and cardiovascular indices in human studies. Growth Horm IGF Res. 2015;25:59–65. doi: 10.1016/j.ghir.2014.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Pedersen BK, Bruunsgaard H. Possible beneficial role of exercise in modulating low-grade inflammation in the elderly. Scand J Med Sci Sports. 2003;13:56–62. doi: 10.1034/j.1600-0838.2003.20218.x. [DOI] [PubMed] [Google Scholar]
- 135.Pedersen BK, Steensberg A, Fischer C, Keller C, Keller P, Plomgaard P, et al. The metabolic role of IL-6 produced during exercise: is IL-6 an exercise factor? Proc Nutr Soc. 2004;63:263–267. doi: 10.1079/PNS2004338. [DOI] [PubMed] [Google Scholar]
- 136.Fillipas S, Cherry CL, Cicuttini F, Smirneos L, Holland AE. The effects of exercise training on metabolic and morphological outcomes for people living with HIV: a systematic review of randomised controlled trials. HIV Clin Trials. 2010;11:270–282. doi: 10.1310/hct1105-270. [DOI] [PubMed] [Google Scholar]
- 137.Fitch K, Abbara S, Lee H, Stavrou E, Sacks R, Michel T, et al. Effects of lifestyle modification and metformin on atherosclerotic indices among HIV-infected patients with the metabolic syndrome. AIDS. 2012;26:587–597. doi: 10.1097/QAD.0b013e32834f33cc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Lindegaard B, Hansen T, Hvid T, van Hall G, Plomgaard P, Ditlevsen S, et al. The effect of strength and endurance training on insulin sensitivity and fat distribution in human immunodeficiency virus-infected patients with lipodystrophy. J Clin Endocrin Metab. 2008;93:3860–3869. doi: 10.1210/jc.2007-2733. [DOI] [PubMed] [Google Scholar]
- 139.Troseid M, Ditlevsen S, Hvid T, Gerstoft J, Grondahl T, Pedersen BK, et al. Reduced trunk fat and triglycerides after strength training are associated with reduced LPS levels in HIV-infected individuals. J Acquir Immune Defic Syndr. 2014;66:e52–54. doi: 10.1097/QAI.0000000000000132. [DOI] [PubMed] [Google Scholar]
- 140.Hu H, Moon J, Chung JH, Kim OY, Yu R, Shin MJ. Arginase inhibition ameliorates adipose tissue inflammation in mice with diet-induced obesity. Biochem Biophys Res Commun. 2015;464:840–847. doi: 10.1016/j.bbrc.2015.07.048. [DOI] [PubMed] [Google Scholar]
- 141.d'Ettorre G, Ceccarelli G, Giustini N, Serafino S, Calantone N, De Girolamo G, et al. Probiotics Reduce Inflammation in Antiretroviral Treated, HIV-Infected Individuals: Results of the “Probio-HIV” Clinical Trial. PLoS One. 2015;10:e0137200. doi: 10.1371/journal.pone.0137200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Stiksrud B, Nowak P, Nwosu FC, Kvale D, Thalme A, Sonnerborg A, et al. Reduced Levels of D-dimer and Changes in Gut Microbiota Composition after Probiotic Intervention in HIV-infected Individuals on Stable ART. J Acquir Immune Defic Syndr. 2015 doi: 10.1097/QAI.0000000000000784. [DOI] [PubMed] [Google Scholar]
- 143.Villar-Garcia J, Hernandez JJ, Guerri-Fernandez R, Gonzalez A, Lerma E, Guelar A, et al. Effect of probiotics (Saccharomyces boulardii) on microbial translocation and inflammation in HIV-treated patients: a double-blind, randomized, placebo-controlled trial. J Acquir Immune Defic Syndr. 2015;68:256–263. doi: 10.1097/QAI.0000000000000468. [DOI] [PubMed] [Google Scholar]
- 144.Vesterbacka J, Barqasho B, Haggblom A, Nowak P. Effects of Co-Trimoxazole on Microbial Translocation in HIV-1-Infected Patients Initiating Antiretroviral Therapy. AIDS Res Hum Retroviruses. 2015;31:830–836. doi: 10.1089/AID.2014.0366. [DOI] [PubMed] [Google Scholar]
- 145.Perez-Matute P, Perez-Martinez L, Aguilera-Lizarraga J, Blanco JR, Oteo JA. Maraviroc modifies gut microbiota composition in a mouse model of obesity: a plausible therapeutic option to prevent metabolic disorders in HIV-infected patients. Rev Esp Quimioter. 2015;28:200–206. [PubMed] [Google Scholar]