Abstract
Over the past 4 decades, the clinical care of people living with HIV (PLWH) evolved from treatment of acute opportunistic infections to the management of chronic, noncommunicable comorbidities. Concurrently, our understanding of adipose tissue function matured to acknowledge its important endocrine contributions to energy balance. PLWH experience changes in the mass and composition of adipose tissue depots before and after initiating antiretroviral therapy, including regional loss (lipoatrophy), gain (lipohypertrophy), or mixed lipodystrophy. These conditions may coexist with generalized obesity in PLWH and reflect disturbances of energy balance regulation caused by HIV persistence and antiretroviral therapy drugs. Adipocyte hypertrophy characterizes visceral and subcutaneous adipose tissue depot expansion, as well as ectopic lipid deposition that occurs diffusely in the liver, skeletal muscle, and heart. PLWH with excess visceral adipose tissue exhibit adipokine dysregulation coupled with increased insulin resistance, heightening their risk for cardiovascular disease above that of the HIV-negative population. However, conventional therapies are ineffective for the management of cardiometabolic risk in this patient population. Although the knowledge of complex cardiometabolic comorbidities in PLWH continues to expand, significant knowledge gaps remain. Ongoing studies aimed at understanding interorgan communication and energy balance provide insights into metabolic observations in PLWH and reveal potential therapeutic targets. Our review focuses on current knowledge and recent advances in HIV-associated adipose tissue dysfunction, highlights emerging adipokine paradigms, and describes critical mechanistic and clinical insights.
Keywords: HIV, ART, adipose tissue dysfunction
Graphical Abstract
Graphical Abstract.
Essential Points.
Antiretroviral therapy (ART) increases longevity in people living with HIV (PLWH) but heightens their risk for a severe form of insulin resistance and metabolic syndrome that increases morbidity and mortality
PLWH manifest excessive ectopic adipose tissue that is most apparent in the liver, heart, and skeletal muscle, ultimately leading to end-organ damage from type 2 diabetes, non-alcoholic fatty liver disease, atherosclerotic cardiovascular disease, and heart failure
The severity and progression of cardiometabolic comorbidities among PLWH are greater than those without HIV, which reflects the complex interplay between HIV persistence in reservoirs and ART effects
HIV viral proteins and ART exposure dysregulate adipose tissue secretion of hormones and microRNAs required for inter-organ crosstalk and metabolic homeostasis
Future efforts to identify clinically meaningful, noninvasive biomarkers of adipose tissue alterations in PLWH will facilitate clinical monitoring and provide important steps toward understanding metabolic comorbidities that currently lack therapeutic options
Adipose tissue (AT) is an endocrine organ that stores energy in lipids and regulates whole-body energy homeostasis via secretion of metabolites and adipokines. To regulate whole-body energy homeostasis, AT adjusts its size and function in response to internal and external stimuli, such as nutritional status, temperature, or changes in energy demands (eg, infection). AT stores triglycerides and, as a result, partitions lipids and lipid derivatives from the periphery. To that end, healthy AT requires the extracellular matrix to expand tissue size (1) and secrete proteins for interorgan crosstalk. In response to excess energy supply, AT expansion leads to adipocyte hypertrophy (increase in size of existing adipocytes) and, in some cases, adipocyte hyperplasia (increase in the number of adipocytes) (2).
Maladaptive AT expansion arises from chronic AT inflammation (3, 4), dysregulation of mitochondrial biogenesis and oxidative phosphorylation (5), excessive extracellular matrix accumulation, fibrosis and hypoxia (6), and altered adipokine secretion, most of which occur in people living with HIV (PLWH) on both older and newer antiretroviral therapy (ART) regimens. PLWH also harbor ectopic AT that is most apparent in the liver, heart, and skeletal muscle. The occurrence of ectopic AT in obesity may be explained by expandability theories that state that ectopic fat depots arise when the capacity to store excess carbon as lipid in subcutaneous and traditional visceral AT depots is exceeded (7). Ectopic fat deposition ultimately associates strongly with end-organ damage, manifested as non-alcoholic fatty liver disease (NAFLD) or atherosclerotic cardiovascular disease (ASCVD). The severity and progression of these metabolic comorbidities and complications are greater in PLWH compared with individuals without HIV. Furthermore, the increased severity of AT dysfunction among PLWH likely contributes to the 3-fold increase in the global burden of HIV-associated cardiovascular disease over the past 2 decades, which is responsible for 2.6 million disability-adjusted life-years per year (8).
Early in the HIV epidemic, PLWH developed severe AT dysfunction characterized by loss of peripheral AT (lipoatrophy), increased central adiposity, insulin resistance, and dyslipidemia (9). These severe metabolic alterations were observed following treatment with the early nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) and protease inhibitors (PIs) (10). With the near-universal discontinuation of toxic ART regimens, the incidence of traditional ART-induced lipodystrophy is now negligible, although lipoatrophy among patients previously exposed to early ART combinations persists as a legacy effect. PLWH treated with integrase strand transfer inhibitors (INSTIs) experience greater weight gain on ART initiation than PLWH treated with non-NRTI (NNRTI) agents. Wasting was not uncommon in the pre- and early-ART eras. The current recommendation for first-line ART among most PLWH is a combination of INSTI and contemporary NRTIs (11, 12), and in the context of this treatment regimen, some PLWH exhibit a “return-to-health” overshoot characterized by excessive AT expansion with unhealthy adipocyte hypertrophy (13, 14). The molecular mechanisms of INSTI-induced weight gain remain incompletely understood but likely derive from metabolic pathways that favor adipocyte hypertrophy, hypoxia, and fibrosis (15). This review expands on contemporary roles for AT in energy homeostasis in the context of HIV-associated metabolic comorbidities. In addition, we discuss current knowledge regarding HIV-associated AT dysfunction and identify important knowledge gaps that challenge attempts to reduce the clinical burden of metabolic disease in PLWH.
AIDS-associated Wasting and Lipodystrophy Reflect Alterations in Adipose Tissue Depots
Before the introduction of effective combination ART in the 1990s, AIDS was common among PLWH. AIDS was associated with multiple endocrine deficiencies, including nonthyroidal illness and adrenal insufficiency in patients with malignancy or opportunistic infections (16). Wasting was perhaps the most devastating metabolic manifestation of AIDS (17, 18) and was defined by weight loss >10% or <90% of ideal body weight (19, 20). Importantly, changes in body composition occurred differently in men than women, with men experiencing a disproportionate decrease in lean body mass with relative fat-sparing, whereas women experienced progressive declines in body fat (21).
Futile cycling emerged as a likely mechanism for AIDS wasting (22, 23). Studies in PLWH and lipodystrophy demonstrated high rates of AT lipolysis coupled with blunted oxidation of the released fatty acids, resulting in increased hepatic flux of the fatty acids to undergo reesterification into triglycerides, secretion from the liver, and repartitioning in AT (24, 25). Increased resting energy expenditure (REE) was an alternate hypothesis for AIDS wasting (17, 20, 26). Although increased REE can be present early in the course of HIV and further increase with progression to AIDS, most patients did not experience significant short-term weight loss despite a significant increase in REE (22). Other studies reported a poorly explained association between GH deficiency (GHD) and AIDS-associated wasting (27, 28).
After the introduction of effective ART, PLWH experienced changes in body fat distribution commonly characterized as mixed lipodystrophy, sometimes termed HIV-associated adipose redistribution syndrome or HIV-associated lipodystrophy syndrome. Mixed lipodystrophy comprised both lipohypertrophy (excessive fat deposition in visceral AT (VAT), dorsocervical region, trunk and/or breasts) and lipoatrophy (characterized by decreased subcutaneous AT [SAT] in face, limbs, and buttocks) (29, 30). Lipoatrophy was quantitatively defined by loss of ≥30% limb fat as measured by dual-energy X-ray absorptiometry, which correlated well with clinically evident lipoatrophy (31). In PLWH and clinical lipodystrophy, dual-energy X-ray absorptiometry and abdominal computed tomography (CT) demonstrate marked peripheral fat loss with moderately increased VAT and relatively preserved lean body mass (32). Lipoatrophy is associated with a cluster of metabolic defects including hypertriglyceridemia, hypercholesterolemia, low high-density lipoprotein (HDL) cholesterol, insulin resistance, lactic acidemia, and steatohepatitis (9, 32, 33).
In addition to exposure to the early PIs and NRTIs (34), other factors contributing to HIV-associated lipoatrophy include age, direct effects of HIV proteins, and a complex but unique dysregulation of lipid kinetics (34). Although the prevalence of lipoatrophy has decreased over time because of improved knowledge of risk factors and the availability of less toxic ART, exposure to some of the early agents (particularly the thymidine analog NRTIs; Table 1) is associated with persistent lipoatrophy after drug cessation, known as a legacy effect (35). Two studies documented a biphasic trend in thymidine analog NRTI-associated lipoatrophy in PLWH, with initial fat gain during the initial 24 to 32 weeks (the “return to health” that was regarded as normal) (35, 36), followed by limb fat loss that varied from 1.7% per year for zidovudine (ZDV)/lamivudine to 19% per year for stavudine/didanosine (35). Of note, thymidine analog NRTI-induced lipoatrophy, although persistent, is partially reversible after switching to a nonthymidine analog NRTI (37). Currently, both the World Health Organization and the US Department of Health and Human Services recommend an INSTI in combination with nonthymidine analog NRTIs as the first-line ART regimen for most PLWH (11, 12).
Table 1.
Adipose tissue redistribution and metabolic endpoints by ART group
| Drug class | Generation | Drug | |
|---|---|---|---|
| Nucleoside reverse transcriptase inhibitors (NRTIs) | Early ART era | Stavudine (D4T)ab | Strongest independent factor associated with lipoatrophy (19%/year fat limb loss when combined with DDI) (35). Associated with type 2 diabetes incidence after adjusting for traditional risk factors and lipodystrophy (38). |
| Zidovudine (ZDV/AZT) and lamivudine (3TC)a | ZDV/3TC combo led to 1.7% limb fat loss (35). Type 2 diabetes incidence persisted after adjusting for traditional risk factors and lipodystrophy (38). Combination ZDV/3TC also decreased insulin-mediated glucose disposal whereas fasting lipolysis was increased (39). |
||
| Didanosine (DDI)b | Incidence of type 2 diabetes incidence remained after adjusting for traditional risk factors and lipodystrophy (38). | ||
| Contemporary ART era | Tenofovir disoproxil fumarate (TDF) | Weight-suppressive effects (40). | |
| Tenofovir alafenamide (TAF) | TDF to TAF switch induced weight gain of 1.8 to 4.47 kg/year regardless of INSTI use (41). TDF to TAF switch is associated with increased LDL and TG levels during the first 9 to 16 months of transition (42). |
||
| Abacavir (ABC) | Higher myocardial infarction risk (43-45). | ||
| Emtricitabine (FTC) | FTC exposure was associated with the lowest T2D risk (adjusted hazard ratio-AHR: 0.30) compared with ABC (AHR: 0.58) and tenofovir (AHR: 0.48) (46). | ||
| Nonnucleoside reverse transcriptase inhibitors (NNRTIs) | Contemporary ART era | Efavirenz (EFV) | EFV increased triglyceride levels (47) coupled with weight-suppressive effects (40). Mean total cholesterol, HDL, LDL, and triglyceride levels increased by 38, 10, 21, and 40 mg/dL in the EFV group after 96 weeks of ART initiation in a prespecified subanalysis of the STARTMRK study of treatment-naïve PLWH (47). |
| Rilpivirine (RPV) | EFV to RPV switch led to decreased serum lipid profiles at weeks 24 and 48 and a significant increase in LDL/HDL ratio at week 48 compared with baseline (48). | ||
| Doravirine (DOR) | ACTG A5391 (phase 4, 48-week, open-label, randomized clinical trial) will evaluate the effect of INSTI/TAF to DOR (and NNRTI) switch plus TAF or TDF in obese adults with HIV, to reduce or prevent further weight gain (49). | ||
| Nevirapine (NVP) | Switching from the indinavir (PI) to NVP decreased insulin levels but did not alter subcutaneous adipocyte apoptosis markers in people with ART-associated lipodystrophy (50). | ||
| Protease inhibitors (PIs) | Early ART era | Indinavir (IDV)b | IDV to NVP switch decreased insulin levels but did not alter subcutaneous adipocyte apoptosis in patients with ART-associated lipodystrophy (50). |
| Ritonavir (RTV) | In men without HIV, 4 weeks of lopinavir (LPV)/RTV increased triglyceride levels, VLDL cholesterol, and FFA levels. 2-hour glucose tolerance was reduced, but no change in insulin-mediated glucose disposal rate was observed (51). | ||
| Saquinavir (SQV)b | SQV and ATV, in combo with TDF/FTC, had comparable modest effects on lipids and little effect on glucose metabolism (52). | ||
| Contemporary ART era | Atazanavir (ATV) | Risk of diabetes development was 27% higher among people who started ART on PIs vs NNRTIs, and similar to INSTI (53). | |
| Darunavir (DRV) | Relative to other EFV-based regimens, DRV/TDF/FTC was associated with an adjusted mean weight gain of 3.68 kg (54). | ||
| Integrase inhibitors (INSTIs) | Contemporary ART era | Dolutegravir (DTG) | Greater weight gain compared with EFV (55, 56). ADVANCE controlled trial suggested a synergistic effect between DTG and TAF on weight gain (55, 57). NA-ACCORD observed a 7.2-kg mean weight gain after 24 months and comparatively beneficial total cholesterol-to-HDL ratio, suggestive of minimal lipid-associated cardiovascular risk among PLWH on INSTIs (58). Although INSTI use was associated with an obesity odds ratio of 1.63, the REPRIEVE cohort reported INSTI use was not associated with differences in glucose, LDL cholesterol, or higher odds of metabolic syndrome or hypertension (59). |
| Bictegravir (BIC) | 4- to 6-kg weight gain vs non-INSTI ART (57, 58). | ||
| Raltegravir (RAL) | Risk of insulin resistance with RAL was similar to that of RTV-boosted ATV or DRV (60). Mean total cholesterol, HDL, and LDL levels increased by 10, 3, and 7 mg/dL in the RAL group, whereas the mean change in total cholesterol, HDL, and LDL levels was 38, 10, and 21 mg/dL in the EFV group after 96 weeks of ART initiation (47). |
||
| Elvitegravir (EVG) | Mean weight gain of 2.72 kg after 96 weeks (57). The mean weight gain was 4.1 kg after 24 months in NA-ACCORD (58). | ||
| Cabotegravir (CAB) | Among PLWH switching to CAB + RPV, changes in weight, BMI, and body composition measurements were minor and similar between treatment arms through months 11-12. There were no clinically relevant changes in the proportion of participants with metabolic syndrome, abdominal obesity, or insulin resistance between arms at months 11-12 (61). |
“Early ART era” denotes ART that were Food and Drug Administration-approved between 1987 and 1997.
Abbreviation: ART, antiretroviral therapy.
a Thymidine analogs.
b No longer available in the United States, Canada, and/or Europe.
Uptake of 18F-fluorodeoxyglucose demonstrated higher metabolic activity of SAT in the extremities and abdomen of ART-treated adult PLWH with lipoatrophy compared with people without HIV. In contrast, 18F-fluorodeoxyglucose uptake in VAT remains unchanged despite increased depot size (62). Additionally, abdominal SAT biopsies from individuals with HIV-associated lipoatrophy demonstrate reduced expression of genes encoding mitochondrial proteins and adipocyte-specific markers compared with persons without HIV, regardless of ART (63). In addition, mRNA expression levels of adiponectin and leptin are downregulated in PLWH compared with persons without HIV, regardless of ART treatment. Taken together, these findings suggest gene expression changes that predict AT competence are present in PLWH before ART initiation (63).
Circulating levels of leptin were noted to be low in PLWH with lipoatrophy and lipohypertrophy (64). This finding motivated a pilot study of treatment with recombinant methionyl leptin (metreleptin), which improved insulin sensitivity and HDL cholesterol levels (65) and decreased VAT (66). Another study reported that metreleptin improved glucose metabolism and non-HDL cholesterol levels but failed to improve abnormal fasting lipid kinetics, triglyceride, or HDL-cholesterol levels in PLWH with lipoatrophy and low fasting leptin levels (≤4 ng/mL) (Table 2) (67). The modest response to metreleptin might be explained by the fact that PLWH exhibit low-normal leptin levels, as opposed to overt leptin deficiency. Despite some beneficial responses in PLWH, metreleptin is not approved for treating HIV lipodystrophy. In fact, metreleptin is only available through a restricted program under a risk evaluation and mitigation strategy (Myalept REMS Program) for the management of congenital or acquired generalized lipodystrophy because of the risks of developing anti-metreleptin antibodies and lymphoma (68).
Table 2.
Randomized controlled trials for HIV-associated lipoatrophy and lipohypertrophy
| Reference | Study population | Intervention | Duration | Outcome |
|---|---|---|---|---|
| Supplements | ||||
| Sutinen et al (69) | PLWH (85% men) with lipoatrophy on ART (n = 10) and people without HIV (n = 10) | Uridine (36 g 3 times daily) | 3 months | Increased total limb fat, intraabdominal fat, and total body fat from baseline. |
| Benedini et al (70) | PLWH on PI/NRTI (n = 9) and people without HIV (n = 9) | Acetyl-L-carnitine (2 g/day) | 8 months | Higher lipid oxidation, decreased intramyocellular triglyceride content, redistribution of fat depots. |
| Calmy et al (71) | Men living with HIV and lipoatrophy | Uridine (36 g 3 times daily) | 24 weeks | No significant change in limb mass. |
| Pharmacologic therapies | ||||
| Grinspoon et al (72) | Eugonadal men living with HIV-associated wasting (n = 54) | Testosterone enanthate (200 mg/week) or placebo injections and progressive resistance training (3 times weekly) or no training | 12 weeks | Muscle mass in limbs improved after testosterone. HDL was significantly decreased. The authors concluded supervised exercise effectively increased muscle mass without the detrimental metabolic effects of testosterone. |
| Falutz et al (73) | PLWH (86% men) with accumulation of abdominal fat (n = 412) | Tesamorelin (2 mg subcutaneous daily) or placebo | 26 weeks | VAT assessed by CT decreased 15.2% in the tesamorelin group and increased by 5.0% in the placebo group. Triglycerides decreased by 50 mg/dL and increased by 9 mg/dL in the placebo group. The ratio of total cholesterol to HDL cholesterol decreased by 0.31 in the tesamorelin group and increased by 0.21 in the placebo group. |
| Slama et al – ANRS 113 (74) | PLWH with lipoatrophy (n = 64) and people without HIV (n = 66) | Pioglitazone 30 mg daily or placebo | 48 weeks | Improved limb fat atrophy in PLWH receiving ART, but clinical benefits were not perceived by the patients. Treatment did lead to a favorable lipid profile. |
| Sekhar et al (67) | Men living with HIV, lipodystrophy, and fasting plasma leptin concentration ≤4 ng/mL (n = 17) | Metreleptin (0.02 mg/kg/day for 2 months, followed by 0.04 mg/kg/day from month 2 to 4) or matching placebo subcutaneously twice daily | 4 months | No differences in BMI, total fat mass, or lean body mass between the study groups at baseline, or after 2 or 4 months of treatment. Two months of treatment with 0.02 or dose escalation to 0.04 mg/kg/day for 2 additional months of metreleptin did not significantly alter rates of total lipolysis, net lipolysis, plasma fatty acid oxidation, or intrahepatic re-esterification. |
Abbreviations: ART, antiretroviral therapy; BMI, body mass index; CT, computed tomography; HDL, high-density lipoprotein; NRTI, nucleoside/nucleotide reverse transcriptase inhibitor; PI, protease inhibitor; PLWH, people living with HIV; VAT, visceral adipose tissue.
Excessive VAT in lipodystrophy impairs GH secretion through increased serum free fatty acids (FFAs) and insulin resistance (75). In contrast, decreasing FFA levels with a potent niacin-derived agent improved GHRH-stimulated secretion of GH in PLWH and lipodystrophy, but not in PLWH without lipodystrophy (75). GHD in PLWH is best discriminated by a serum GH peak <7.5 mg/L after provocative testing with GHRH plus arginine (76). Based on this cutoff point, the prevalence of GHD among PLWH is estimated to be approximately 36.9 per 100 000 adults (76). By comparison, the incidence of adult-onset GHD among individuals without HIV ranges between 1.42 (females) to 1.9 (males) per 100 000 adults (77). The factors influencing abnormal basal and stimulated GH secretion patterns in PLWH include age, gender, body composition, VAT mass, and intrinsic HIV factors, and have been reviewed elsewhere (78).
PLWH Exhibit a “Return to Health” Overshoot in the Contemporary ART Era
Weight gain is a complex physiological phenomenon modified by age, gender, ethnicity, diet, and activity. In the contemporary ART era, up to 65% of PLWH may be classified as overweight or obese in resource-limited and resource-abundant settings (79-81). Weight gain following ART initiation is accompanied by both beneficial and detrimental outcomes. For example, the Veterans Aging Cohort Study reported improved survival with weight gain within the first year of ART in PLWH who started treatment when normal-weight or underweight, whereas overweight or obese individuals lacked a clinical benefit (82), consistent with a return-to-health effect. although a return to normal weight may be desired in underweight persons and a small amount of weight gain in overweight or obese persons may be tolerable after ART initiation, some PLWH demonstrate weight gain that exceeds what could reasonably reflect a “return to health.” Excessive weight gain beyond a “return to health” state has been observed more frequently in PLWH treated with INSTIs and the NRTI tenofovir alafenamide (TAF) (Table 1) (57). One hypothesis suggests that a favorable gastrointestinal side effect profile might partly explain greater weight gain with newer ART (57).
Although PIs are not part of the recommended first-line ART regimen, it is worth noting the effects of PIs on weight gain. In the Centers for AIDS Research Network of Integrated Clinical Systems cohort, 6-month weight gain after ART initiation was evaluated in 3232 treatment-naïve participants (84% men). Darunavir (DRV) in combination with tenofovir disoproxil fumarate (TDF)/emtricitabine (FTC) induced a mean weight gain of 3.68 kg during the initial 6 months of ART in treatment-naïve individuals when compared with efavirenz (EFV)/TDF/FTC (54). This weight gain was only surpassed by dolutegravir (DTG) (mean weight gain, 4.37 kg) and bictegravir (BIC) (mean weight gain, 3.86 kg) in combination with TAF/FTC and persisted after adjusting for multiple factors (54). Of note, both TDF and EFV may have weight-suppressive effects (40), complicating data interpretation of cumulative effects of ART on body composition and weight.
The ADVANCE trial, a phase 3, open-label, randomized clinical trial compared the combination of FTC and DTG plus TAF or TDF against the standard-of-care regimen EFV/TDF/FTC. After 48 weeks, persons randomized to DTG-based ART gained significantly more weight than persons randomized to EFV (mean increase, 6.4 kg in the DTG/TAF group, 3.2 kg in the DTG/TDF group, and 1.7 kg in the standard-of-care group), with the greatest weight gain seen in participants treated with DTG/TAF. These trends in weight gain continued to week 96 without a plateau in DTG-treated persons. Weight gain was significantly higher in females in all 3 groups (55). As previously mentioned, TDF and EFV may also have weight-suppressive effects (40). However, in most studies, the difference between TDF and TAF is only about 2 to 3 kg (41, 83, 84), making the clinical relevance of differences between the 2 tenofovir prodrugs unclear. Numerous other ART start-and-switch studies support the findings of the ADVANCE trial (58, 85). Results from a large US observational database (OPERA) demonstrated accelerated weight gain from 1.8 to 4.47 kg/year following TDF to TAF switch, regardless of whether PLWH switched from a boosted PI or NNRTI to an INSTI (Table 1), suggesting an independent effect of TAF on weight gain (41). There were differences among the different INSTIs. Combinations of BIC/TAF, DTG/TAF, or elvitegravir/cobicistat/TAF induced 4.47 kg/year, 3.09 kg/year, and 2.55 kg/year weight gain, respectively, within the first 9 months following the switch from non-INSTI to INSTI. However, after the initial 9 months, there was no significant additional weight change attributable to TAF (41).
In the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) trial, current INSTI use was associated with higher odds of obesity (odds ratio = 1.63), mean body mass index (BMI) change (+1.5 kg/m2), and larger waist circumference (+3.6 cm) compared with non-INSTI drugs. Differences in weight-related to INSTI use were greater among women and non-White participants, with sex and race additively affecting BMI (59). Importantly, INSTI use was not associated with differences in fasting blood glucose, low-density lipoprotein (LDL) cholesterol, or odds of metabolic syndrome or hypertension (59). In contrast, the large North American HIV cohort (NA-ACCORD) reported a type 2 diabetes hazard ratio of 1.17 in the INSTI group that was attenuated to 1.03 after adjusting for 12-month weight gain (53).
Although some efforts have been undertaken to explore the mechanisms of ART-induced weight gain, the causal effects of ART and HIV on body composition remain incompletely understood. In mice, a high-fat diet admixed with ART (EFV/FTC/TDF) additively increased pro-inflammatory genes in VAT and impaired glucose metabolism and energy balance (86). In vitro exposure of adipose stem cells to DTG was associated with greater lipid accumulation via elevated expression of pro-adipogenic and lipogenic transcription factors (87). Moreover, DTG and raltegravir (RAL) decreased expression and secretion of adiponectin and leptin (87). In the same study, larger adipocytes were observed in the SAT and VAT of macaques treated with DTG (87). Other in vitro studies showed that DTG significantly reduced brown adipogenic markers, such as uncoupling protein-1 (UCP1) (88). Moreover, DTG reduced UCP1 and levels of mitochondrial complex IV components, which contributed to depleted mitochondrial respiratory capacity and slowed insulin-stimulated glucose uptake (88). DTG and BIC also decreased expression of the brown adipocyte markers including Cbp/P300-interacting-transactivator-1 (CITED1), PR domain containing 16 (PRDM16), and iodothyronine deiodinase-2 (DIO2) (15). Mechanistically, DTG induced hypoxia-inducible factor 1 subunit alpha (HIF1A) expression and elevated fibrosis markers (15), which strongly correlated with repressed PRDM16, UCP1, and DIO2. Altogether, a few animal and cell-based studies suggest INSTI-induced weight gain occurs because of cellular metabolic programs that favor adipocyte hypertrophy, hypoxia, and fibrosis (15).
AT Dysfunction Contributes to Ectopic Fat Accumulation in PLWH
HIV-associated Nonalcoholic Fatty Liver Disease
NAFLD is an independent risk factor for cardiovascular disease, type 2 diabetes, and all-cause mortality (89). NAFLD occurs on a well-defined disease spectrum ranging from steatosis (increased hepatic triglyceride content) to non-alcoholic steatohepatitis (NASH) with or without fibrosis to advanced irreversible scarring (cirrhosis), hepatocellular carcinoma, and end-stage liver disease (90, 91). NAFLD affects 25% of adults worldwide (92). In contrast, the pooled prevalence of HIV-associated NAFLD and significant fibrosis are estimated at 38% (95% CI, 31-45) and 13% (95% CI, 8-18), respectively (93).
In a case-control study comparing age- and sex-matched patients with primary NAFLD (n = 33) and patients with HIV-associated NAFLD (n = 33), patients with HIV-associated NAFLD had significantly higher mean aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, and serum triglycerides (94). Similarly, compared with age and sex-matched primary NAFLD, patients with HIV-associated NAFLD had significantly higher rates of steatohepatitis (37% vs 63%, P = .04), and features of liver injury, including lobular inflammation and acidophil bodies (94). Although obesity is associated with NAFLD in the general population, NAFLD is noted in PLWH at significantly lower mean BMI, suggesting PLWH manifest higher prevalence of “lean NAFLD” (Fig. 1) (95).
Figure 1.
Characteristics of primary NAFLD vs HIV-associated NAFLD. Primary NAFLD is associated with obesity in the general population, whereas HIV-associated NAFLD is noted at a significantly lower mean BMI. In a head-to-head comparison between age- and sex-matched people with primary and HIV-associated NAFLD, people with HIV-associated NAFLD showed significantly higher mean liver enzymes, alkaline phosphatase, and serum triglycerides. Similarly, patients with HIV-associated NAFLD had significantly higher rates of biopsy-proven steatohepatitis and features of liver injury, including lobular inflammation and acidophil bodies.
The natural progression of HIV-associated NAFLD was assessed in a subanalysis of a double-blind trial that evaluated the effects of tesamorelin vs placebo on NAFLD (96). In the placebo group, fibrosis progressed in 38% (n = 9) of participants with HIV-associated NAFLD over a 12-month period (97). Furthermore, 5 of 9 participants with fibrosis progression had no evidence of fibrosis at baseline. Baseline VAT was higher among volunteers with fibrosis progression than without progression (306 ± 119 cm2 vs 212 ± 89 cm2; P = .04). In multivariable regression modeling, a 25 cm2 higher VAT at baseline was associated with a 37% increased odds of fibrosis progression on adjusting for baseline NAFLD Activity Score, liver fat content, and BMI (odds ratio, 1.37; 95% CI, 1.03-2.07; P = .03). Of note, 50% (n = 12) of subjects had no change in fibrosis and 13% (n = 3) experienced fibrosis regression (97).
The high prevalence of NAFLD in PLWH was previously ascribed to coinfection with hepatitis C virus (HCV) or adverse ART effects, particularly those of stavudine (98-103). However, several epidemiological studies reported that only 50% of HIV-associated liver diseases were attributable to hepatitis B virus and HCV coinfection or alcohol abuse (104-106). Moreover, new data demonstrate that PLWH develop NAFLD and accelerated liver disease without HCV or hepatitis B virus coinfection (98). The factors that promote development and progression of a specific, HIV-associated form of NAFLD are likely multifactorial and closely tied to AT dysfunction. Greater VAT accumulation predicts risk for NAFLD and metabolic syndrome in PLWH (97, 107). Kinetic studies using stable isotope infusions and mass spectrometry identified accelerated AT lipolysis and defective whole-body oxidation of fatty acids as an important element in the pathophysiology of NAFLD in PLWH (24, 25), associated with a mixed hyperlipidemia in which triglycerides, very low LDL and LDL are increased (108). Additionally, HIV-associated hormonal imbalances such as hypogonadism or increased renin-angiotensin system activation appear to contribute to energy balance regulation and possibly NAFLD risk in PLWH (109, 110).
As previously mentioned, early ART agents (NRTIs and PIs) were associated with a lipodystrophic syndrome of which NAFLD is a component (9, 111). Older NRTIs may contribute to NAFLD pathogenesis through altered mitochondrial toxicity, insulin resistance, and redistribution of AT to the visceral space (112, 113). Moreover, both NRTIs and NNRTIs affect adipogenesis and adipocyte differentiation, leading to mitochondrial injury, adipocyte death, and FFA and triglyceride accumulation (113), which correlate with hepatic steatosis. Contributions of specific drugs to NAFLD risk in the current ART era are under investigation but likely mediate risk in part through their effects on weight gain and body composition.
Because there are no US Food and Drug Administration-approved medications for NAFLD treatment, management includes lifestyle changes that promote weight loss of at least 5% (preferably ≥10% weight loss) because weight loss is associated with greater liver and cardiometabolic benefit (114). Consideration of weight loss and insulin-sensitizing (eg, pioglitazone) medications are recommended in the general population (114). Few NAFLD/NASH treatment trials have included PLWH. Therapy options for HIV-associated NAFLD may include pioglitazone because this medication was well-tolerated and reduced hepatic fat by magnetic resonance spectroscopy in individuals with HCV and HIV coinfection (115). An open-label clinical trial recently showed vitamin E decreases markers of liver fat content and hepatocyte apoptosis in PLWH with biopsy-proven NASH (116). The GHRH analog tesamorelin improves liver enzymes, hepatic steatosis, and angiogenic, fibrogenic, and pro-inflammatory mediators compared with placebo (96, 117, 118). The effectiveness of glucagon-like peptide-1 receptor agonists for management of NAFLD in PLWH is under study (119).
Skeletal Muscle Fat Accumulation
Intermuscular adipose tissue (IMAT) accumulates beneath the muscle fascia and between bundles of myocytes. Both VAT and IMAT increase with age, whereas SAT and skeletal muscle mass decrease (120, 121). The Fat Redistribution and Metabolic Change in HIV Infection study was the first large cross-sectional study to assess the total IMAT in 425 PLWH and 211 individuals without HIV from diverse ethnic groups (122). Contrary to predictions, the Fat Redistribution and Metabolic Change in HIV Infection researchers found the total IMAT assessed by magnetic resonance imaging was 51% lower in participants with HIV compared with participants without HIV (P = .003), a finding that was attenuated but persisted in multivariate analyses (122). When stratified by sex, IMAT was significantly lower in men but not in women. The authors attributed the lower IMAT to prior exposure to NRTIs such as stavudine, didanosine, and nelfinavir (122). A recent study by Adrian et al demonstrated that the PI DRV/ritonavir was associated with greater fat area and lower density of both fat and muscle, and RAL with less intermuscular fat in the psoas (123). The clinical relevance of IMAT relies on its resemblance to VAT, which constitutes a marker of increased metabolic and cardiovascular disease risk in the general population (124, 125), and metabolic syndrome among PLWH (126). The relationship between metabolic and cardiovascular health and improvements in VAT or other fat depots is not straightforward. PLWH exhibited improvements in VAT after 1 year of lifestyle modifications but experienced an increase in LDL and significant progression of carotid intima-media thickness (127), a surrogate marker for cardiovascular disease. These findings underscore the complex relationships between AT maladaptation and cardiometabolic risk in PLWH.
Intramyocardial and Epicardial Fat Accumulation
HIV intramyocardial steatosis is associated with diastolic dysfunction (128, 129), a precursor of heart failure with both preserved ejection fraction and reduced ejection fraction (130), both of which are prevalent among PLWH (131, 132). In healthy conditions, cardiomyocytes populating the myocardial structural space contain approximately 0.4% to 0.5% triglycerides (133, 134). The REPRIEVE trial reported a median intramyocardial triglyceride content of 0.59% (Q1, Q3: 0.28%, 1.15%). Intramyocardial triglyceride content (IMTC) was increased (>0.5%) among 52% and markedly increased (>1.5%) among 22% of participants (135). Parameters associated with increased myocardial steatosis included age, BMI ≥ 25 kg/m2, history of IV drug use, and CD4+ T-cell count < 350 cells/mm3 (135). Importantly, 67% of individuals with increased IMTC (>0.5%), 71% with nonmarkedly increased IMTC (≤1.5%), and 72% with markedly increased IMTC (>1.5%) had a normal waist (<102 cm in men, <88 cm in women) (135). These results suggest that intramyocardial steatosis in PLWH occurs even in individuals without abdominal obesity.
Men living with HIV also accumulate epicardial fat, which is associated with a higher risk of cardiovascular events compared with men without HIV with similar cardiovascular and metabolic disease risk profiles (136). Ectopic epicardial fat volume in HIV is associated with VAT area, fasting glucose, and insulin. The associations with elevated blood glucose and insulin remained significant after controlling for age, race, BMI, adiponectin, VAT, and ART (137). Recently, epicardial fat volume was shown to correlate with abnormal cardiac structure and function as evidenced by left ventricular mass index and left ventricular global longitudinal strain, respectively (138). Furthermore, Guaraldi et al showed an association between epicardial fat and traditional risk factors for atherosclerosis, suggesting epicardial fat may be a useful marker of cardiovascular risk in PLWH (139). Because the sequelae of expanding epicardial and intramyocardial fat depots resemble those of NAFLD and its increased cardiometabolic risk, intrinsic (viral proteins) and extrinsic (ART) HIV-specific factors likely generate parallel effects in driving these ectopic fat depots, including triggering proinflammatory and profibrotic changes at the cellular level. These findings underscore the importance of ectopic fat accumulation as a target for the management of cardiometabolic alterations in PLWH.
Comorbidities of Obesity Contribute to Additional Disease Burden in PLWH
Lipotoxicity, Insulin Resistance, and Type 2 Diabetes in PLWH
In 2019, the crude incidence of type 2 diabetes (T2D) was estimated at 5.9 per 1000 among diagnosed US adults aged 18 years or older (140). By comparison, the incidence of T2D and prediabetes among PLWH has been estimated to be 13.7 per 1000 person-years and 125 per 1000 person-years, respectively (141). The increased incidence of T2D among PLWH is likely related to increased circulating levels of lipids that contribute to lipotoxicity in the peripheral organs, which in turn leads to insulin resistance in the liver and skeletal muscle (142). Lipotoxicity is characterized by the accumulation of lipolysis mediators (eg, palmitate derivatives), which may account for the hypertriglyceridemia seen in lipodystrophic PLWH (24, 25). To date, no published studies have addressed the role of lipotoxicity on insulin resistance or T2D in the contemporary ART era. In contrast, most efforts have been directed toward addressing the role of AT inflammation as a mechanism for insulin resistance and T2D in PLWH. Indeed, several studies reported that HIV and simian immunodeficiency virus (SIV) are associated with a substantial increase in the proportion of adipose CD8+ T cells relative to CD4+ T cells, which is remarkably similar to enrichments of T cells described in obesity (143-145).
A study that compared 8 non- and prediabetic HIV-negative persons and 9 (of 26) PLWH paired by similar age, glycated hemoglobin, and BMI reported that, despite a much higher proportion of CD8+ T cells in the SAT of PLWH, there was no significant difference in the overall proportions of SAT CD4+ and CD8+ memory T-cell subsets between PLWH and HIV-negative persons (146). Of note, there were significant gender (10% vs 63% females, P = .02) and race (40% vs 100% Caucasian, P < .01) differences between the HIV subgroup and non-HIV controls that could potentially explain the lack of differences between groups (146). Thus, the contribution of AT inflammation to insulin resistance and T2D in PLWH remains uncertain. In addition, these results could also explain why treatment with medications that diminish vascular inflammation, such as statins, show conflicting results in PLWH (147, 148).
Significant efforts were also devoted to assessing the contribution of different ART regimens on the risk of T2D or prediabetes, as this was a modifiable factor compared with aging, family history of diabetes, or Black or Hispanic origin (141). In the early ART era, the main T2D risk factor in PLWH was PI use. Indeed, incidence rates of hyperglycemia as high as 5-fold were reported in the setting of PI use (149). Mechanisms that account for these effects include skeletal muscle and AT insulin resistance, and β-cell impairment in PLWH exposed to 12 weeks of PI (150). However, the effects of PIs on glucose homeostasis vary widely among the different PIs. For example, an in vitro study demonstrated that an oligopeptide found in indinavir inhibited glucose transport in primary rat adipocytes via reversible noncompetitive inhibition of GLUT4 (151), whereas a clinical study of PLWH showed atazanavir in combination with ritonavir had a modest effect on insulin sensitivity compared with lopinavir/ritonavir, as measured by glucose disposal rate during hyperinsulinemic-euglycemic clamps (152). Moreover, at therapeutic doses, atazanavir does not inhibit glucose uptake in vitro or in vivo (153, 154).
In addition to PIs, older-generation NRTIs have been associated with T2D (Table 1) (38). In contrast to PIs, the mechanism for NRTI-induced T2D was attributed to mitochondrial dysfunction. One study found that 1-month treatment with stavudine was associated with decreased mitochondrial DNA from muscle biopsies and insulin sensitivity, as measured by the glucose infusion rate during hyperinsulinemic-euglycemic clamps (155). As previously mentioned, thymidine analog NRTIs were the main culprit of lipoatrophy; hence, lipolysis was increased by 22% after 3 months of combination lamivudine/ZDV (39). The risk of T2D for stavudine, ZDV, and DDI persisted after adjusting for traditional risk factors and lipodystrophy (38). Similar to the newer generation of PIs, the use of newer NRTIs, including FTC, abacavir, and tenofovir, is associated with a lower risk of T2D (46).
Case reports of new-onset diabetes have been reported in conjunction with INSTI use (156). However, this finding has not been replicated in larger studies. Despite its class effect on weight gain, INSTIs are not associated with differences in fasting blood glucose, LDL cholesterol, or odds of metabolic syndrome or hypertension (59). Furthermore, the NA-ACCORD reported a T2D hazard ratio of 1.17 in the INSTI group that was attenuated to 1.03 after adjusting for 12-month weight gain (Table 1) (53). One clinical trial compared RAL with 2 boosted PI regimens and found that the increases in homeostasis model assessment-insulin resistance in all 3 arms were not significantly different from one another and appeared to be independent of changes in VAT (60).
Multiple observational studies shed light on important associations between different risk factors, cellular mediators and markers, and metabolic phenotypes in PLWH. Further mechanistic studies will be necessary to dissect individual contributions of HIV viral proteins, ART regimens, and chronic immune activation to the pathophysiology of insulin resistance in the contemporary ART era. Only then will we be able to devise new therapeutic strategies for managing these HIV-associated metabolic comorbidities and limit their impact on the quality of life of PLWH.
ASCVD Burden in PLWH
Metabolic abnormalities are tightly associated with ASCVD risk factors in adults with HIV (157). The disproportionate incidence of cardiometabolic comorbidities and complications negatively affects the quality of life of this population. Large cohort studies demonstrated a high prevalence of hypertension, dyslipidemia, and insulin resistance resulting from greater VAT among PLWH (126). The high prevalence of known cardiovascular risk factors only partially accounts for the 3-fold ASCVD risk in PLWH compared with individuals without HIV (8). Furthermore, conventional therapies (eg, statins) exert modest effects for primary or secondary prevention of ASCVD in PLWH (32).
ASCVD risk prevention is based on disease prediction using individual risk factors. Traditional ASCVD risk factors such as age, T2D, current smoking, hypertension, and dyslipidemia are associated with elevated ASCVD risk among PLWH, as in the general population (158, 159). HIV constitutes an independent ASCVD risk factor (158), especially in individuals with low current or nadir CD4+ T-cell counts or a history of sustained untreated HIV (160). Although no general population risk assessment models focused on PLWH, the latest American Heart Association scientific statement on management of ASCVD in HIV recommends applying the ACC/ASCVD risk estimator, Framingham cardiovascular disease risk estimator, or D:A:D plus risk enhancer factors, such as history of prolonged HIV viremia or delayed ART initiation, low current or nadir CD4+ T-cell count (<350 cell/mm3), HIV treatment failure or nonadherence, metabolic syndrome/lipoatrophy/lipohypertrophy, NAFLD, or hepatitis C co-infection (161).
Associations between intra-abdominal VAT and increased metabolic and cardiovascular disease risk are well described in cross-sectional and longitudinal studies (124, 125). Large cohort studies demonstrate a higher prevalence of hypertension, dyslipidemia, and insulin resistance resulting from greater VAT among PLWH (126). Despite a reduction in VAT after 1 year of lifestyle modifications (diet, exercise, smoking cessation, and antidiabetic/dyslipidemic therapy), PLWH with and without lipodystrophy, exhibited an increase in LDL and had significant progression of carotid intima-media thickness (127), a surrogate marker for cardiovascular disease in the general population.
Taken together, the role of AT dysfunction in ASCVD risk in PLWH cannot be predicted from changes in immune cells or fat distribution. Intricate interactions between diverse cellular mediators, including circulating metabolites from AT, may play a more prominent role in the ASCVD risk in PLWH.
PLWH Exhibit Intestinal Dysbiosis That Corresponds With AT Inflammation
The gastrointestinal tract is colonized extensively with commensal microorganisms collectively called the microbiota, which play an important role in metabolism and immune defense as part of a symbiotic relationship with the host. Dysbiosis depicts an imbalance in the microbiota composition that frequently accompanies the chronic inflammation observed in generalized obesity (162) and PLWH (163). A few studies reported intestinal dysbiosis and reduction of bacterial alpha diversity in PLWH compared with individuals without HIV (164, 165). Not surprisingly, PLWH accumulate markers of bacterial translocation and immune activation that correspond with inflammation in AT during acute and chronic infections. Bacterial alpha diversity correlates with VAT and systemic inflammation (166), strengthening the notion that intestinal microbiota contribute to ectopic fat accumulation in PLWH.
Bacteria within the gut break down dietary fibers and starch through anaerobic fermentation, producing short-chain fatty acids as metabolite byproducts. Although short-chain fatty acids maintain intestinal homeostasis, they quickly enter the bloodstream and can be metabolized by peripheral tissues including AT (167). In addition to loss of gut microbial diversity, obesity and HIV are associated with lower butyrate-producing bacteria (168). The potential relationships between visceral obesity, gut microbiota, short-chain fatty acid abundance, and immune activation in people living with HIV are an active area of research, including the potential roles of dysbiosis in altering ART outcomes.
AT Stress Alters Secretion of Metabolic Hormones That Change Metabolism in PLWH
Efficient interorgan communication facilitates metabolic adaptation to energy demands. In addition to its role in nutrient sensing and energy homeostasis, AT secretes a large number of bioactive molecules (eg, adipokines, microRNAs [miRNAs]) that participate in immune responses and metabolic regulation through their paracrine and/or endocrine actions (169). In addition, adipose-derived extracellular vesicles carry miRNAs and proteins, which act as cellular messengers for interorgan crosstalk from fat cells. The communication between AT and other organs is altered in many pathological conditions, such as obesity, diabetes, and HIV. However, only a few candidate molecules that perform interorgan communication have been rigorously studied in PLWH.
Leptin
The identification of the adipocyte-specific protein hormone leptin defined AT as a major endocrine organ (170) that signals energy deficit to the central nervous system (171). Leptin modulates energy homeostasis (172) and counteracts metabolic dysfunction during overnutrition by increasing lipolysis and glucose uptake and inhibiting hepatic glucose output (173). Leptin physiology in PLWH is variably disrupted. Some PLWH and lipoatrophy exhibit low leptin levels because of reduced adipocyte mass, whereas others exhibit high leptin levels, suggesting leptin resistance (174). Conversely, in a larger clinical trial (AIDS Clinical Trials Group A5260s), mean leptin levels increased 22% after 96 months of ART initiation, whereas mean adiponectin changed 1% (175). Furthermore, higher circulating leptin was associated with greater homeostasis model assessment-insulin resistance and high-sensitivity C-reactive protein independent of fat depot size, suggesting that greater adipocyte lipid content contributed to impaired glucose tolerance and systemic inflammation among PLWH starting ART (175). Possibly because of the variable leptin levels among PLWH, treatment with metreleptin yielded modest results. PLWH exhibit low-normal leptin levels, as opposed to overt leptin deficiency, which may explain modest effects of metreleptin in clinical trials (65-67). However, as noted previously, metreleptin is not approved for management of lipodystrophy in PLWH.
Adiponectin
Adiponectin targets diverse tissues and generates insulin-sensitizing, antiatherogenic, and anti-inflammatory properties (176, 177). In addition, adiponectin enhances hepatic fatty acid oxidation, suppresses hepatic de novo lipogenesis (178), and increases ceramidase activity, thus preventing or reversing diet-induced steatosis, insulin resistance, and glucose intolerance (179). The role of adiponectin as a marker of metabolic health in PLWH is not clear. In a cardiovascular substudy of the Multicenter AIDS Cohort Study, higher adiponectin levels were associated with metabolic health in men with HIV (180). Conversely, in the same cohort, lower adiponectin levels were observed in men with and without HIV and were associated with coronary stenosis by CT angiography (181). Furthermore, low adiponectin levels in men living with HIV correlate with the severity of subclinical atherosclerosis, independent of traditional cardiovascular disease risk factors (181). In contrast, another study reported a correlation between higher adiponectin levels and worse neuropsychological test scores in men with HIV, suggesting that adiponectin excess may contribute to cognitive decline in men living with HIV. Adiponectin levels in this study were inversely correlated with VAT (r = −0.37, P < .0001) and positively correlated with thigh SAT (r = 0.19, P < .001) area in men with HIV, suggesting a disequilibrium of traditional relationships between AT, adipokines, and end-organ damage (182). Moreover, lean ART-treated men with HIV displayed low adiponectin levels that mirror those of obese, insulin-resistant men without HIV (183).
Adiponectin changes following ART vary (184, 185). For example, among treatment-naïve PLWH, adiponectin increased by 9%, 8%, and 1% in persons treated with ATV/r, DRV/r, and RAL, respectively, after 48 weeks (186). Conversely, a decline in adiponectin from baseline to 48 weeks was associated with a rise in high-sensitivity C-reactive protein and IL-6 (175). Some studies have tried to clarify the role of leptin and adiponectin in PLWH by estimating the leptin/adiponectin ratio, a surrogate biomarker of metabolic competence in the general population (187). Interestingly, a small single-center study in Japan reported a significant increase in leptin/adiponectin ratio and serum leptin levels 12 months after initiation of DTG- or BIC-based ART (188). These results may reflect the increased size of AT depots observed on INSTI therapy initiation.
Plasminogen-activator Inhibitor 1
Plasminogen-activator inhibitor 1 (PAI-1) is another protein that can be secreted in abundance by VAT in obese people and predicts insulin resistance (189) and NAFLD (190, 191). In large epidemiological studies including the Framingham study, serum PAI-1 levels have been associated with increased fasting glucose (192), systolic blood pressure, and triglyceride levels (193). Similarly, PAI-1 appears to be an independent predictor of impaired insulin sensitivity (194) and fatty liver disease in PLWH with lipodystrophy (195). However, conflicting results have also been reported in PLWH. One study reported that PAI-1 expression was significantly reduced at the mRNA and protein level in PLWH compared with matched persons without HIV, a finding that appeared to be related to NNRTI exposure (196). In contrast, in lipodystrophic PLWH, plasma PAI-1 levels positively correlated with BMI (r = 0.74, P < .01), TNF-α level (r = 0.64, P < .01), and VAT quantity (r = 0.67, P < .01) (197). Cytokines such as TNF-α, IL-6, and IL-8 are determinants of adiponectin and PAI-1 abundance in plasma, underscoring the role of inflammation on adipokines and cardiometabolic risk in PLWH (196, 197).
Fibroblast Growth Factor 21
Secretion of the protein hormone fibroblast growth factor 21 (FGF21) from the liver improves glucose clearance and insulin sensitivity (198, 199). In healthy mice and people, the most prompt and consistent effects of an acute rise in serum FGF21 levels are increased fat oxidation and lipolysis suppression in concert with insulin (200-202). Along these lines, FGF21 is required for acute insulin sensitization of AT, manifested by enhanced glucose uptake, blunted lipolysis, and reduced plasma FFAs (203). Although FGF21 physiology is well-documented, we do not yet understand how FGF21 contributes to the metabolic phenotype of PLWH.
Despite observations that FGF21 is secreted from AT and exerts paracrine effects (204), FGF21 is primarily secreted from the liver (205). Circulating FGF21 levels are increased in PLWH compared with controls without HIV, regardless of ART (206-208), but this is associated with an unhealthy metabolic profile and precursor lesions for liver cancer. FGF21 levels in PLWH strongly and positively correlate with lipodystrophy, dyslipidemia, and NAFLD (206). FGF21 levels also correlate with the predictors of hepatocellular carcinoma, hepatic steatosis, and NASH in other conditions of altered energy metabolism, such as obesity (209). Identifying clinically meaningful relationships between FGF21 and chronic liver disease will enhance our understanding of the pathogenesis of hepatocellular cancer risk in PLWH, facilitate clinical monitoring, and potentially provide a noninvasive endpoint for future therapeutic studies.
FGF21 appearance in the context of fatty liver disease likely reflects energetic stress on peripheral tissues and consequently lacks physiological impacts on glucose metabolism (210, 211). FGF21 increases thermogenic gene expression, including UCP1, in specific white adipose tissue depots (212). Srinivasa et al noted that FGF21 in PLWH is associated with increased expression of brown adipose tissue marker genes in subcutaneous dorsocervical fat biopsies (208). These observations argue that FGF21 resistance occurs in specific tissues, such as AT, that renders elevated levels of this hormone unable to reverse metabolic defects.
Growth Differentiating Factor 15
Growth differentiation factor 15 (GDF15) is a recently acknowledged circulating peptide that exerts weight-lowering effects when administered to rodents and nonhuman primates (213, 214). Intracellular stress drives the expression of GDF15 in the AT, kidney, liver, skeletal muscle, and some immune cells. Other cellular factors that elevate GDF15 include mitochondrial dysfunction (215-219), the integrated stress response (216, 220), and AMP-associated protein kinase activation (221). Consequently, GDF15 expression is upregulated in chronic inflammatory diseases (222), cardiovascular disease (218, 223), NAFLD (224), and diabetes (218, 223, 225).
As with FGF21, the activation of transcription downstream of the integrated stress response drives expression of GDF15. Specific HIV-related risk factors (eg, viral proteins such as viral protein R [Vpr], immune activation, ART) may result in concerted stress that alters the course of liver disease and sustained GDF15 secretion. Men living with HIV have higher plasma GDF15 levels than HIV-negative men (both healthy and with metabolic syndrome) (226). There was a positive correlation between GDF15 levels and age in healthy subjects and subjects with metabolic syndrome, but not in PLWH, which may reflect premature aging in PLWH.
Glial cell line-derived neurotrophic factor receptor family receptor alpha-like, a transmembrane receptor localized to the hindbrain (227) binds GDF15 and mediates its satiety effects in rodents (220). In PLWH, higher GDF15 levels are associated with greater weight loss after 12-week treatment with metformin (228). However, it remains unknown whether GDF15 induces appetite suppression effects in this population. Additional studies will be needed to test whether GDF15 levels in PLWH are associated with fatty liver disease and resistance to the central nervous effects of GDF15, similar to other hormone resistance paradigms in PLWH (229). Nonetheless, the satiety and weight loss effects of GDF15 represent important biological endpoints to slow weight gain in PLWH and manage HIV-associated metabolic comorbidities.
microRNAs
microRNAs (miRNAs) are noncoding RNAs of 20 to 25 nucleotides that bind target mRNAs in the 3′ untranslated region to induce mRNA degradation and inhibit protein translation (230). DICER is an integral protein in the miRNA processing required for adipocyte functions (231). Mice lacking Dicer in adipocytes (Dicer a-KO) display lipodystrophy, insulin resistance, fatty liver, and premature mortality (231). Similarly, PLWH and lipodystrophy exhibit reductions in AT Dicer expression (232). These observations strongly support a rate-limiting role for miRNAs in the functions of adipose tissue in PLWH.
AT-derived exosomal miRNAs likely represent a new form of adipokine. In addition to their endogenous actions, miRNAs can be transported into the extracellular space within nanoparticles (50-150 nm in size) termed exosomes. Exosomes transport most of the miRNA secretome and allow paracrine and endocrine transfer of miRNA silencing into circulation. Dicer a-KO depletes circulating miRNAs that communicate with peripheral tissues (233). Along these lines, transplantation of wild-type white adipose tissue into Dicer a-KO restored plasma miRNAs and improved glucose tolerance. Dicer a-KO also showed reduced miR-99b in circulating exosomes coupled with higher liver Fgf21 mRNA, which could be partially corrected with the administration of miR-99b loaded into exosomes. These mechanisms of Fgf21 regulation derived from AT exosomes may be relevant for interpreting the hyperelevation of plasma FGF21 observed in PLWH.
Furthermore, expression of many circulating miRNAs correlated significantly to adipose Dicer expression among all participants (persons without HIV; PLWH with and without lipodystrophy). Moreover, the lipodystrophic miRNA signature strongly correlated with dorsocervical AT area (a clinical sign of lipodystrophy) and decreased expression of the brown adipocyte genes, UCP1 and PRM16 (234). Taken together, these data suggest that alterations at the level of circulating miRNAs from AT serve as markers of lipodystrophy among PLWH. More studies will be needed to understand the endocrine functions and predictive power of miRNAs in AT exosomes in PLWH.
Preclinical Models Define Adipose Tissue as a Reservoir for Latent HIV and Viral Proteins
Preclinical Models for HIV-associated Metabolic Dysfunction
Animal models have provided critical insights into HIV latency and reservoirs (Fig. 2). Although many models have been developed to study HIV/AIDS (235), very few proved to be effective for the study of cardiometabolic consequences. Whereas humanized mice make it possible to model HIV infection of human cells in vivo and design studies using genetically identical animals, these models are limited in their ability to replicate the effects of HIV and ART on nonhematopoietic tissues and to recreate basic features of HIV disease in humans (235).
Figure 2.
Adipose tissue is an important anatomic HIV reservoir. The main cellular reservoirs for HIV are CD4+ T cells and macrophages. After acute HIV infection, surviving HIV-infected CD4+ T cells and macrophages continue to harbor and sustain the proliferation of provirus and viral proteins. ART initiation (dashed line) decreases new viral replication and cellular infection but does not eliminate the survival of these latently infected cells, which can persist in anatomical reservoirs, such as AT. In aviremic PLWH on ART, CD4+ T cells of AT harbor replication-competent HIV with a median latent HIV proviral DNA copy number equivalent to that of circulating and mesenteric lymph node CD4+ T cells. Ultimately, HIV viral proteins and ART exposure contribute to chronic AT inflammation and excessive extracellular matrix accumulation.
Rhesus macaques with SIV infection are the most prevalent animal models used to study HIV pathogenesis. These models provided critical insights into the physiopathology of HIV-associated metabolic complications. The AT of SIV-infected rhesus macaques harbors infected immune cells early during infection, with subsequent defects in AT metabolism (236). Moreover, SIV DNA and RNA are present in AT stromal vascular fraction cells, CD4+ T cells, and CD14+ macrophages in SAT and VAT from SIV-infected macaques (237). Furthermore, in vitro cultures of AT stromal vascular cells from PLWH and macaques demonstrated the limited antiviral effectiveness of some NRTIs, particularly tenofovir, whereas the INSTI DTG penetrates AT and effectively inhibits viral replication (Fig. 2) (238). The main limitation of rhesus macaque models is the high costs of colony maintenance.
Mouse models helped formalize the notion that factors released by HIV provoke a confluence of chronic maladaptive responses in AT that result in dyslipidemia and insulin resistance. Transgenic mouse models for HIV accessory Vpr (239) and negative factor (Nef) (240) provided critical knowledge about virus-driven mechanisms of metabolic disease in PLWH. Subsequently, recombinant Vpr and Nef were developed, allowing in vivo studies via continuous intraperitoneal infusions (241, 242). The use of adenovirus-associated virus to expand the nature of viral protein expression may provide new tools for expressing HIV-specific factors in tissue-specific ways and allow diet, strain, and ART effects to be explored in vivo.
The phenotype of the Vpr mouse models recapitulates the critical aspects of HIV-associated metabolic disease in PLWH (241, 243). Enforced Vpr expression, via transgenic expression or continuous infusion of recombinant Vpr, in the skeletal muscle, liver, brain, and AT upregulates AT lipolysis and impairs adipocyte differentiation, leading to lipoatrophy with AT inflammation (241). In the liver, Vpr expression causes accelerated lipogenesis, impaired fatty acid oxidation, slowed fatty acid export, and steatohepatitis (243). As a result, the mice exhibit glucose intolerance and hypertriglyceridemia. Although mechanisms of Vpr-associated AT dysfunction are incompletely understood, Vpr animal models remain the most viable way to study metabolic complications related to HIV and ART.
The in vivo roles of Tat and Nef are less clear, with studies suggesting both inhibition and facilitation of adipocyte differentiation (244-246). Although the contributions of specific HIV viral proteins to metabolic derangement in PLWH remain to be fully delineated, the mechanistic data from experimental animal models and the presence of Vpr, Tat, and Nef in the plasma of PLWH on suppressive ART strongly suggest a causal role for HIV viral proteins in AT dysfunction.
AT is an Anatomic HIV Reservoir
Data from animal models set the foundation for experiments that defined HIV persistence in anatomic reservoirs of PLWH. ART inhibits HIV viral replication and partially restores immune function but does not fully eradicate HIV virions, leading to persistence in cellular and anatomic reservoirs. AT is an important reservoir for HIV harboring activated memory CD4+ T cells and HIV DNA, even in the setting of suppressive ART (Fig. 2) (247). In aviremic PLWH on ART, replication-competent HIV is present in CD4+ T cells of both SAT and VAT with a median latent HIV proviral DNA copy number equivalent to that of circulating and mesenteric lymph node CD4+ T cells (237). These data indicate that AT supports viral persistence and chronic immune activation and inflammation in ART-treated PLWH (Fig. 2).
Conclusions
PLWH manifest excessive ectopic AT that is most apparent in the liver, heart, and skeletal muscle and ultimately leads to end-organ damage from T2D, NAFLD, ASCVD, and heart failure. The increased severity of AT dysfunction among PLWH likely contributes to the 3-fold increase in the global burden of HIV-associated cardiovascular disease over the past 2 decades (8).
We lack a fundamental understanding of why ART and viral factors alter metabolism in PLWH because animal models are limited. In the transgenic Vpr mouse model, expression of the HIV accessory protein Vpr is sufficient to cause all the cardinal manifestations of HIV-associated metabolic disease (226, 241, 243, 248). Thus, the Vpr animal models are the most viable way to study metabolic complications related to HIV and ART. Animal models remain the most important preclinical tools for testing medical interventions and acquiring the basic scientific knowledge that will ultimately be needed to develop safe therapies to manage the long-term metabolic impact of HIV and ART. Thus, additional animal models and methods using contemporary gene editing and transgenic approaches (249) may enable the study of HIV-related risk factors that contribute to metabolic diseases. However, a critical limitation of rodent models in the study of immune activation in HIV resides in the biological differences between human and rodent gut barrier integrity, one of the underlying factors that mediate chronic inflammation even in virologically suppressed PLWH.
HIV-related factors and ART likely contribute to metabolic deterioration in PLWH, but the endocrine arms of these responses remain unclear. Although many biomarkers have been explored, the limitations of study design do not draw a clear picture, and interpretations are limited to purely correlative observations. Reports of inflammatory markers that go up or down in the serum of PLWH also abound. Emergent biomarkers derived from AT may also be relevant for predicting metabolic disease outcomes in PLWH. AT is a major contributor to the circulating small RNAs carried by small extracellular vesicles, which have regulatory functions in distant tissues (250). Although AT secretes extracellular vesicles that uniquely reflect lipodystrophy in PLWH (233, 234) and likely mediate responses in recipient cells and tissues, more studies will be needed to understand their endocrine functions and predictive power.
Significant pitfalls must be considered to identify valid mechanistic biomarkers in PLWH, including uniformity in measurement techniques and confounding factors related to age, BMI, and sex differences. For example, insulin resistance has been associated with lower serum adiponectin and higher triglycerides among men living with HIV and with hyperleptinemia among women living with HIV, respectively (251). In persons without HIV, NAFLD prevalence is higher in men than in premenopausal women, whereas NAFLD tends to become more common in women after menopause (252). Estrogen (253) may engender the differences in metabolic syndrome prevalence and liver cancer risk between men and women living with HIV. Unfortunately, most of the available data in PLWH comes disproportionately from studies of men living with HIV (254). Hence, the impact of sex hormones on HIV comorbidities might be underestimated. Considering that women and minorities living with HIV have a greater prevalence of obesity and are at greater risk of experiencing poorer outcomes from HIV treatment (255, 256), there is a clear need for biomarker studies of NAFLD and disease risk in these communities. Analysis of large cohort data, such as the MACS/WIHS Combined Cohort Study, with contemporary methodologies (eg, high-throughput sequencing and multi-omics approaches), will be needed to define and clarify biomarkers that predict metabolic disease in PLWH.
Abbreviations
- ASCVD
atherosclerotic cardiovascular disease
- ART
antiretroviral therapy
- AT
adipose tissue
- BIC
bictegravir
- BMI
body mass index
- CT
computed tomography
- DICER a-KO
lacking Dicer in adipocytes
- DRV
darunavir
- DTG
dolutegravir
- EFV
efavirenz
- FFA
free fatty acid
- FGF21
fibroblast growth factor 21
- FTC
emtricitabine
- GDF15
growth differentiation factor 15
- GHD
GH deficiency
- HCV
hepatitis C virus
- HDL
high-density lipoprotein
- IMAT
intermuscular adipose tissue
- IMTC
intramyocardial triglyceride content
- INSTI
integrase strand transfer inhibitor
- LDL
low-density lipoprotein
- miRNA
microRNA
- NAFLD
non-alcoholic fatty liver disease
- NASH
non-alcoholic steatohepatitis
- Nef
negative factor
- NNRTI
non-nucleoside/nucleotide reverse transcriptase inhibitor
- NRTI
nucleoside/nucleotide reverse transcriptase inhibitor
- PAI-1
plasminogen activator inhibitor-1
- PI
protease inhibitor
- PLWH
people living with HIV
- RAL
raltegravir
- REE
resting energy expenditure
- SAT
subcutaneous adipose tissue
- SIV
simian immunodeficiency virus
- T2D
type 2 diabetes
- TAF
tenofovir alafenamide
- TDF
tenofovir disoproxil fumarate
- VAT
visceral adipose tissue
- Vpr
viral protein R
- ZDV
zidovudine
Contributor Information
Claudia E Ramirez Bustamante, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
Neeti Agarwal, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
Aaron R Cox, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
Sean M Hartig, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
Jordan E Lake, Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA.
Ashok Balasubramanyam, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
Funding
This work was funded by the National Institutes of Health grants 5T32AI055413 (C.E.R.B), R01DK126042 (J.E.L), and R01DK114356 (S.M.H). C.E.R.B is also supported by the Baylor St. Luke's Roderick D. MacDonald Fellowship. A.B. is supported by the Rutherford Fund for Diabetes Research.
Disclosures
J.E.L. serves as a consultant to Theratechnologies. The remaining authors declared no conflicts of interest.
References
- 1. Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clement K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 2019;129(10):4032‐4040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Jo J, Gavrilova O, Pack S, et al. Hypertrophy and/or hyperplasia: dynamics of adipose tissue growth. PLoS Comput Biol. 2009;5(3):e1000324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutrients and inflammation. J Clin Invest. 2008;118(9):2992‐3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Czech MP. Insulin action and resistance in obesity and type 2 diabetes. Nat Med. 2017;23(7):804‐814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Chouchani ET, Kajimura S. Metabolic adaptation and maladaptation in adipose tissue. Nat Metab. 2019;1(2):189‐200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Sun K, Halberg N, Khan M, Magalang UJ, Scherer PE. Selective inhibition of hypoxia-inducible factor 1α ameliorates adipose tissue dysfunction. Mol Cell Biol. 2013;33(5):904‐917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Virtue S, Vidal-Puig A. It’s not how fat you are, it’s what you do with it that counts. PLoS Biol. 2008;6(9):e237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Shah ASV, Stelzle D, Lee KK, et al. Global burden of atherosclerotic cardiovascular disease in people living with HIV: systematic review and meta-analysis. Circulation. 2018;138(11):1100‐1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Carr A, Samaras K, Burton S, et al. A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors. AIDS. 1998;12(7):F51‐F58. [DOI] [PubMed] [Google Scholar]
- 10. Vigouroux C, Gharakhanian S, Salhi Y, et al. Adverse metabolic disorders during highly active antiretroviral treatments (HAART) of HIV disease. Diabetes Metab. 1999;25(5):383‐392. [PubMed] [Google Scholar]
- 11. World Health Organization . Updated recommendations on first-line and second-line antiretroviral regimens and post-exposure prophylaxis and recommendations on early infant diagnosis of HIV. Accessed February 3, 2022. https://www.who.int/publications/i/item/WHO-CDS-HIV-18.51
- 12. Department of Health and Human Services. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. Accessed January 3, 2022. https://clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-arv
- 13. Peckham SC, Entenman C, Carroll HW. The influence of a hypercaloric diet on gross body and adipose tissue composition in the rat. J Nutr. 1962;77(2):187‐197. [DOI] [PubMed] [Google Scholar]
- 14. Steinberg MD, Zingg W, Angel A. Studies of the number and volume of fat cells in adipose tissue. J Pediatr. 1962;61(2):299‐300. [DOI] [PubMed] [Google Scholar]
- 15. Ngono Ayissi K, Gorwood J, Le Pelletier L, et al. Inhibition of adipose tissue beiging by HIV integrase inhibitors, dolutegravir and bictegravir, is associated with adipocyte hypertrophy, hypoxia, elevated fibrosis, and insulin resistance in simian adipose tissue and human adipocytes. Cells. 2022;11(11):1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Sellmeyer DE, Grunfeld C. Endocrine and metabolic disturbances in human immunodeficiency virus infection and the acquired immune deficiency syndrome. Endocr Rev. 1996;17(5):518‐532. [DOI] [PubMed] [Google Scholar]
- 17. Macallan DC, Noble C, Baldwin C, et al. Energy expenditure and wasting in human immunodeficiency virus infection. N Engl J Med. 1995;333(2):83‐88. [DOI] [PubMed] [Google Scholar]
- 18. Macallan DC. Wasting in HIV infection and AIDS. J Nutr. 1999;129(1S Suppl):238S‐242S. [DOI] [PubMed] [Google Scholar]
- 19. Rivera S, Briggs W, Qian D, Sattler FR. Levels of HIV RNA are quantitatively related to prior weight loss in HIV-associated wasting. J Acquir Immune Defic Syndr Hum Retrovirol. 1998;17(5):411‐418. [DOI] [PubMed] [Google Scholar]
- 20. Melchior JC, Salmon D, Rigaud D, et al. Resting energy expenditure is increased in stable, malnourished HIV-infected patients. Am J Clin Nutr. 1991;53(2):437‐441. [DOI] [PubMed] [Google Scholar]
- 21. Grinspoon S, Corcoran C, Miller K, et al. Body composition and endocrine function in women with acquired immunodeficiency syndrome wasting. J Clin Endocrinol Metab. 1997;82(5):1332‐1337. [DOI] [PubMed] [Google Scholar]
- 22. Grunfeld C, Feingold KR. Metabolic disturbances and wasting in the acquired immunodeficiency syndrome. N Engl J Med. 1992;327(5):329‐337. [DOI] [PubMed] [Google Scholar]
- 23. Grunfeld C. What causes wasting in AIDS? N Engl J Med. 1995;333(2):123‐124. [DOI] [PubMed] [Google Scholar]
- 24. Sekhar RV, Jahoor F, White AC, et al. Metabolic basis of HIV-lipodystrophy syndrome. Am J Physiol Endocrinol Metab. 2002;283(2):E332‐E337. [DOI] [PubMed] [Google Scholar]
- 25. Sekhar RV, Jahoor F, Pownall HJ, et al. Severely dysregulated disposal of postprandial triacylglycerols exacerbates hypertriacylglycerolemia in HIV lipodystrophy syndrome. Am J Clin Nutr. 2005;81(6):1405‐1410. [DOI] [PubMed] [Google Scholar]
- 26. Grunfeld C, Pang M, Shimizu L, Shigenaga JK, Jensen P, Feingold KR. Resting energy expenditure, caloric intake, and short-term weight change in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. Am J Clin Nutr. 1992;55(2):455‐460. [DOI] [PubMed] [Google Scholar]
- 27. Heijligenberg R, Sauerwein HP, Brabant G, Endert E, Hommes MJ, Romijn JA. Circadian growth hormone secretion in asymptomatic human immune deficiency virus infection and acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1996;81(11):4028‐4032. [DOI] [PubMed] [Google Scholar]
- 28. Rondanelli M, Solerte SB, Fioravanti M, et al. Circadian secretory pattern of growth hormone, insulin-like growth factor type I, cortisol, adrenocorticotropic hormone, thyroid-stimulating hormone, and prolactin during HIV infection. AIDS Res Hum Retroviruses. 1997;13(14):1243‐1249. [DOI] [PubMed] [Google Scholar]
- 29. Lo JC, Mulligan K, Tai VW, Algren H, Schambelan M. “Buffalo hump” in men with HIV-1 infection. Lancet. 1998;351(9106):867‐870. [DOI] [PubMed] [Google Scholar]
- 30. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM) . Fat distribution in women with HIV infection. J Acquir Immune Defic Syndr. 2006;42(5):562‐571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Podzamczer D, Ferrer E, Martinez E, et al. How much fat loss is needed for lipoatrophy to become clinically evident? AIDS Res Hum Retroviruses. 2009;25(6):563‐567. [DOI] [PubMed] [Google Scholar]
- 32. Carr A. HIV Lipodystrophy: risk factors, pathogenesis, diagnosis and management. AIDS. 2003;17(Suppl 1):S141‐S148. [PubMed] [Google Scholar]
- 33. Carr A, Miller J, Law M, Cooper DA. A syndrome of lipoatrophy, lactic acidaemia and liver dysfunction associated with HIV nucleoside analogue therapy: contribution to protease inhibitor-related lipodystrophy syndrome. AIDS. 2000;14(3):F25‐F32. [DOI] [PubMed] [Google Scholar]
- 34. Caron-Debarle M, Lagathu C, Boccara F, Vigouroux C, Capeau J. HIV-associated lipodystrophy: from fat injury to premature aging. Trends Mol Med. 2010;16(5):218‐229. [DOI] [PubMed] [Google Scholar]
- 35. Dube MP, Komarow L, Mulligan K, 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(5):508‐514. [DOI] [PubMed] [Google Scholar]
- 36. Mallon PW, Miller J, Cooper DA, Carr A. Prospective evaluation of the effects of antiretroviral therapy on body composition in HIV-1-infected men starting therapy. AIDS. 2003;17(7):971‐979. [DOI] [PubMed] [Google Scholar]
- 37. Martin A, Mallon PW. Therapeutic approaches to combating lipoatrophy: do they work? J Antimicrob Chemother. 2005;55(5):612‐615. [DOI] [PubMed] [Google Scholar]
- 38. De Wit S, Sabin CA, Weber R, et al. Incidence and risk factors for new-onset diabetes in HIV-infected patients: the data collection on adverse events of anti-HIV drugs (D:A:D) study. Diabetes Care. 2008;31(6):1224‐1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Blumer RM, van Vonderen MG, Sutinen J, et al. Zidovudine/lamivudine contributes to insulin resistance within 3 months of starting combination antiretroviral therapy. AIDS. 2008;22(2):227‐236. [DOI] [PubMed] [Google Scholar]
- 40. Glidden DV, Mulligan K, McMahan V, et al. Metabolic effects of preexposure prophylaxis with coformulated tenofovir disoproxil fumarate and emtricitabine. Clin Infect Dis. 2018;67(3):411‐419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Mallon PW, Brunet L, Hsu RK, et al. Weight gain before and after switch from TDF to TAF in a U.S. cohort study. J Int AIDS Soc. 2021;24(4):e25702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Mallon PWG, Brunet L, Fusco JS, et al. Lipid changes after switch from TDF to TAF in the OPERA cohort: LDL cholesterol and triglycerides. Open Forum Infect Dis. 2022;9(1):ofab621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. D:A:D Study Group . Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients enrolled in the D:A:D study: a multi-cohort collaboration. Lancet. 2008;371(9622):1417‐1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Young J, Xiao Y, Moodie EE, et al. Effect of cumulating exposure to abacavir on the risk of cardiovascular disease events in patients from the Swiss HIV cohort study. J Acquir Immune Defic Syndr. 2015;69(4):413‐421. [DOI] [PubMed] [Google Scholar]
- 45. Marcus JL, Neugebauer RS, Leyden WA, et al. Use of abacavir and risk of cardiovascular disease among HIV-infected individuals. J Acquir Immune Defic Syndr. 2016;71(4):413‐419. [DOI] [PubMed] [Google Scholar]
- 46. Spagnuolo V, Galli L, Poli A, et al. Associations of statins and antiretroviral drugs with the onset of type 2 diabetes among HIV-1-infected patients. BMC Infect Dis. 2017;17(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Lennox JL, Dejesus E, Berger DS, et al. Raltegravir versus efavirenz regimens in treatment-naive HIV-1-infected patients: 96-week efficacy, durability, subgroup, safety, and metabolic analyses. J Acquir Immune Defic Syndr. 2010;55(1):39‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Phongsamart W, Jantarabenjakul W, Chantaratin S, et al. Switching efavirenz to rilpivirine in virologically suppressed adolescents with HIV: a multi-centre 48-week efficacy and safety study in Thailand. J Int AIDS Soc. 2022;25(1):e25862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. AIDS Clinical Trials Group. Doravirine for Persons With Excessive Weight Gain on Integrase Inhibitors and Tenofovir Alafenamide. Accessed February 9, 2022. https://www.clinicaltrials.gov/study/NCT04636437?intr=Doravirine%20Tenofovir%20Alafenamide&rank=5
- 50. Domingo P, Matias-Guiu X, Pujol RM, et al. Switching to nevirapine decreases insulin levels but does not improve subcutaneous adipocyte apoptosis in patients with highly active antiretroviral therapy-associated lipodystrophy. J Infect Dis. 2001;184(9):1197‐1201. [DOI] [PubMed] [Google Scholar]
- 51. Lee GA, Seneviratne T, Noor MA, et al. The metabolic effects of lopinavir/ritonavir in HIV-negative men. AIDS. 2004;18(4):641‐649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Vrouenraets SM, Wit FW, Fernandez Garcia E, et al. Randomized comparison of metabolic and renal effects of saquinavir/r or atazanavir/r plus tenofovir/emtricitabine in treatment-naive HIV-1-infected patients. HIV Med. 2011;12(10):620‐631. [DOI] [PubMed] [Google Scholar]
- 53. Rebeiro PF, Jenkins CA, Bian A, et al. Risk of incident diabetes mellitus, weight gain, and their relationships with integrase inhibitor-based initial antiretroviral therapy among persons with human immunodeficiency virus in the United States and Canada. Clin Infect Dis. 2021;73(7):e2234‐e2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Ruderman SA, Crane HM, Nance RM, et al. Brief report: weight gain following ART initiation in ART-naive people living with HIV in the current treatment era. J Acquir Immune Defic Syndr. 2021;86(3):339‐343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Venter WDF, Moorhouse M, Sokhela S, et al. Dolutegravir plus two different prodrugs of tenofovir to treat HIV. N Engl J Med. 2019;381(9):803‐815. [DOI] [PubMed] [Google Scholar]
- 56. Calmy A, Tovar Sanchez T, Kouanfack C, et al. Dolutegravir-based and low-dose efavirenz-based regimen for the initial treatment of HIV-1 infection (NAMSAL): week 96 results from a two-group, multicentre, randomised, open label, phase 3 non-inferiority trial in Cameroon. Lancet HIV. 2020;7(10):e677‐e687. [DOI] [PubMed] [Google Scholar]
- 57. Sax PE, Erlandson KM, Lake JE, et al. Weight gain following initiation of antiretroviral therapy: risk factors in randomized comparative clinical trials. Clin Infect Dis. 2020;71(6):1379‐1389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Bourgi K, Jenkins CA, Rebeiro PF, et al. Weight gain among treatment-naive persons with HIV starting integrase inhibitors compared to non-nucleoside reverse transcriptase inhibitors or protease inhibitors in a large observational cohort in the United States and Canada. J Int AIDS Soc. 2020;23(4):e25484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Kileel EM, Lo J, Malvestutto C, et al. Assessment of obesity and cardiometabolic status by integrase inhibitor use in REPRIEVE: a propensity-weighted analysis of a multinational primary cardiovascular prevention cohort of people with human immunodeficiency virus. Open Forum Infect Dis. 2021;8(12):ofab537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Dirajlal-Fargo S, Moser C, Brown TT, et al. Changes in insulin resistance after initiation of raltegravir or protease inhibitors with tenofovir-emtricitabine: AIDS Clinical Trials Group A5260s. Open Forum Infect Dis. 2016;3(3):ofw174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Tan DHS, Antinori A, Eu B, et al. Weight and metabolic changes with cabotegravir + rilpivirine long-acting or bictegravir. Presented at: CROI; February 19–22, 2023 2023; Seattle, Washington. Session 6. https://www.croiconference.org/abstract/weight-and-metabolic-changes-with-cabotegravirrilpivirine-long-acting-or-bictegravir/
- 62. Hadigan C, Kamin D, Liebau J, et al. Depot-specific regulation of glucose uptake and insulin sensitivity in HIV-lipodystrophy. Am J Physiol Endocrinol Metab. 2006;290(2):E289‐E298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Giralt M, Domingo P, Guallar JP, et al. HIV-1 infection alters gene expression in adipose tissue, which contributes to HIV-1/HAART-associated lipodystrophy. Antivir Ther. 2006;11(6):729‐740. [PubMed] [Google Scholar]
- 64. Nagy GS, Tsiodras S, Martin LD, et al. Human immunodeficiency virus type 1-related lipoatrophy and lipohypertrophy are associated with serum concentrations of leptin. Clin Infect Dis. 2003;36(6):795‐802. [DOI] [PubMed] [Google Scholar]
- 65. Lee JH, Chan JL, Sourlas E, Raptopoulos V, Mantzoros CS. Recombinant methionyl human leptin therapy in replacement doses improves insulin resistance and metabolic profile in patients with lipoatrophy and metabolic syndrome induced by the highly active antiretroviral therapy. J Clin Endocrinol Metab. 2006;91(7):2605‐2611. [DOI] [PubMed] [Google Scholar]
- 66. Mulligan K, Khatami H, Schwarz JM, et al. The effects of recombinant human leptin on visceral fat, dyslipidemia, and insulin resistance in patients with human immunodeficiency virus-associated lipoatrophy and hypoleptinemia. J Clin Endocrinol Metab. 2009;94(4):1137‐1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Sekhar RV, Jahoor F, Iyer D, et al. Leptin replacement therapy does not improve the abnormal lipid kinetics of hypoleptinemic patients with HIV-associated lipodystrophy syndrome. Metabolism. 2012;61(10):1395‐1403. [DOI] [PubMed] [Google Scholar]
- 68. Amylin Pharmaceuticals LLC . Initial REMS approval. Accessed July 3, 2023. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/125390Orig1s000Rems.pdf
- 69. Sutinen J, Walker UA, Sevastianova K, et al. Uridine supplementation for the treatment of antiretroviral therapy-associated lipoatrophy: a randomized, double-blind, placebo-controlled trial. Antivir Ther. 2007;12(1):97‐105. [PubMed] [Google Scholar]
- 70. Benedini S, Perseghin G, Terruzzi I, et al. Effect of L-acetylcarnitine on body composition in HIV-related lipodystrophy. Horm Metab Res. 2009;41(11):840‐845. [DOI] [PubMed] [Google Scholar]
- 71. Calmy A, Bloch M, Wand H, et al. No significant effect of uridine or pravastatin treatment for HIV lipoatrophy in men who have ceased thymidine analogue nucleoside reverse transcriptase inhibitor therapy: a randomized trial. HIV Med. 2010;11(8):493‐501. [DOI] [PubMed] [Google Scholar]
- 72. Grinspoon S, Corcoran C, Parlman K, et al. Effects of testosterone and progressive resistance training in eugonadal men with AIDS wasting. A randomized, controlled trial. Ann Intern Med. 2000;133(5):348‐355. [DOI] [PubMed] [Google Scholar]
- 73. Falutz J, Allas S, Blot K, et al. Metabolic effects of a growth hormone-releasing factor in patients with HIV. N Engl J Med. 2007;357(23):2359‐2370. [DOI] [PubMed] [Google Scholar]
- 74. Slama L, Lanoy E, Valantin MA, et al. Effect of pioglitazone on HIV-1-related lipodystrophy: a randomized double-blind placebo-controlled trial (ANRS 113). Antivir Ther. 2008;13(1):67‐76. [PubMed] [Google Scholar]
- 75. Koutkia P, Meininger G, Canavan B, Breu J, Grinspoon S. Metabolic regulation of growth hormone by free fatty acids, somatostatin, and ghrelin in HIV-lipodystrophy. Am J Physiol Endocrinol Metab. 2004;286(2):E296‐E303. [DOI] [PubMed] [Google Scholar]
- 76. Lo J, You SM, Wei J, Canavan B, Grinspoon S. Relationship of peak growth hormone to cardiovascular parameters, waist circumference, lipids and glucose in HIV-infected patients and healthy adults. Clin Endocrinol (Oxf). 2009;71(6):815‐822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Stochholm K, Gravholt CH, Laursen T, et al. Incidence of GH deficiency - a nationwide study. Eur J Endocrinol. 2006;155(1):61‐71. [DOI] [PubMed] [Google Scholar]
- 78. Rochira V, Guaraldi G. Growth hormone deficiency and human immunodeficiency virus. Best Pract Res Clin Endocrinol Metab. 2017;31(1):91‐111. [DOI] [PubMed] [Google Scholar]
- 79. 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(1):159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Buchacz K, Baker RK, Palella FJ Jr, 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(1):65‐75. [DOI] [PubMed] [Google Scholar]
- 81. Ali MK, Magee MJ, Dave JA, 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‐S39. [DOI] [PubMed] [Google Scholar]
- 82. Yuh B, Tate J, Butt AA, et al. Weight change after antiretroviral therapy and mortality. Clin Infect Dis. 2015;60(12):1852‐1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Gomez M, Seybold U, Roider J, Harter G, Bogner JR. A retrospective analysis of weight changes in HIV-positive patients switching from a tenofovir disoproxil fumarate (TDF)- to a tenofovir alafenamide fumarate (TAF)-containing treatment regimen in one German university hospital in 2015–2017. Infection. 2019;47(1):95‐102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Orkin C, Eron JJ, Rockstroh J, et al. Week 96 results of a phase 3 trial of darunavir/cobicistat/emtricitabine/tenofovir alafenamide in treatment-naive HIV-1 patients. AIDS. 2020;34(5):707‐718. [DOI] [PubMed] [Google Scholar]
- 85. Esber AL, Chang D, Iroezindu M, et al. Weight gain during the dolutegravir transition in the African Cohort Study. J Int AIDS Soc. 2022;25(4):e25899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Pepin ME, Padgett LE, McDowell RE, et al. Antiretroviral therapy potentiates high-fat diet induced obesity and glucose intolerance. Mol Metab. 2018;12:48‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Gorwood J, Bourgeois C, Pourcher V, et al. The integrase inhibitors dolutegravir and raltegravir exert proadipogenic and profibrotic effects and induce insulin resistance in human/simian adipose tissue and human adipocytes. Clin Infect Dis. 2020;71(10):e549‐e560. [DOI] [PubMed] [Google Scholar]
- 88. Jung I, Tu-Sekine B, Jin S, et al. Dolutegravir suppresses thermogenesis via disrupting uncoupling protein 1 expression and mitochondrial function in brown/beige adipocytes in preclinical models. J Infect Dis. 2022;226(9):1626‐1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Ma W, Wu W, Wen W, et al. Association of NAFLD with cardiovascular disease and all-cause mortality: a large-scale prospective cohort study based on UK Biobank. Ther Adv Chronic Dis. 2022;13:20406223221122478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Brunt EM. Pathology of nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol. 2010;7(4):195‐203. [DOI] [PubMed] [Google Scholar]
- 91. Brunt EM, Wong VWS, Nobili V, et al. Nonalcoholic fatty liver disease. Nat Rev Dis Primers. 2015;1(1):15080. [DOI] [PubMed] [Google Scholar]
- 92. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73‐84. [DOI] [PubMed] [Google Scholar]
- 93. Manzano-Nunez R, Rivera-Esteban J, Navarro J, et al. Uncovering the NAFLD burden in people living with HIV from high- and middle-income nations: a meta-analysis with a data gap from Subsaharan Africa. J Int AIDS Soc. 2023;26(3):e26072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Vodkin I, Valasek MA, Bettencourt R, Cachay E, Loomba R. Clinical, biochemical and histological differences between HIV-associated NAFLD and primary NAFLD: a case-control study. Aliment Pharmacol Ther. 2015;41(4):368‐378. [DOI] [PubMed] [Google Scholar]
- 95. Mohammed SS, Aghdassi E, Salit IE, et al. HIV-positive patients with nonalcoholic fatty liver disease have a lower body mass index and are more physically active than HIV-negative patients. J Acquir Immune Defic Syndr. 2007;45(4):432‐438. [DOI] [PubMed] [Google Scholar]
- 96. Fourman LT, Stanley TL, Billingsley JM, et al. Delineating tesamorelin response pathways in HIV-associated NAFLD using a targeted proteomic and transcriptomic approach. Sci Rep. 2021;11(1):10485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Fourman LT, Stanley TL, Zheng I, et al. Clinical predictors of liver fibrosis presence and progression in human immunodeficiency virus-associated nonalcoholic fatty liver disease. Clin Infect Dis. 2021;72(12):2087‐2094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Crum-Cianflone N, Dilay A, Collins G, et al. Nonalcoholic fatty liver disease among HIV-infected persons. J Acquir Immune Defic Syndr. 2009;50(5):464‐473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Gaslightwala I, Bini EJ. Impact of human immunodeficiency virus infection on the prevalence and severity of steatosis in patients with chronic hepatitis C virus infection. J Hepatol. 2006;44(6):1026‐1032. [DOI] [PubMed] [Google Scholar]
- 100. Marks KM, Petrovic LM, Talal AH, Murray MP, Gulick RM, Glesby MJ. Histological findings and clinical characteristics associated with hepatic steatosis in patients coinfected with HIV and hepatitis C virus. J Infect Dis. 2005;192(11):1943‐1949. [DOI] [PubMed] [Google Scholar]
- 101. McGovern BH, Ditelberg JS, Taylor LE, et al. Hepatic steatosis is associated with fibrosis, nucleoside analogue use, and hepatitis C virus genotype 3 infection in HIV-seropositive patients. Clin Infect Dis. 2006;43(3):365‐372. [DOI] [PubMed] [Google Scholar]
- 102. Monto A, Dove LM, Bostrom A, Kakar S, Tien PC, Wright TL. Hepatic steatosis in HIV/hepatitis C coinfection: prevalence and significance compared with hepatitis C monoinfection. Hepatology. 2005;42(2):310‐316. [DOI] [PubMed] [Google Scholar]
- 103. Sulkowski MS, Mehta SH, Torbenson M, et al. Hepatic steatosis and antiretroviral drug use among adults coinfected with HIV and hepatitis C virus. AIDS. 2005;19(6):585‐592. [DOI] [PubMed] [Google Scholar]
- 104. Ferguson TF, Rosen E, Carr R, et al. Associations of liver disease with alcohol use among people living with HIV and the role of hepatitis C: the New Orleans alcohol use in HIV study. Alcohol Alcohol. 2020;55(1):28‐36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Pembroke T, Deschenes M, Lebouche B, et al. Hepatic steatosis progresses faster in HIV mono-infected than HIV/HCV co-infected patients and is associated with liver fibrosis. J Hepatol. 2017;67(4):801‐808. [DOI] [PubMed] [Google Scholar]
- 106. Salloum S, Holmes JA, Jindal R, et al. Exposure to human immunodeficiency virus/hepatitis C virus in hepatic and stellate cell lines reveals cooperative profibrotic transcriptional activation between viruses and cell types. Hepatology. 2016;64(6):1951‐1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Price JC, Ma Y, Kuniholm MH, et al. Human immunodeficiency virus is associated with elevated FibroScan-aspartate aminotransferase (FAST) score. Clin Infect Dis. 2022;75(12):2119‐2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Heeren J, Scheja L. Metabolic-associated fatty liver disease and lipoprotein metabolism. Mol Metab. 2021;50:101238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Bhasin S, Parker RA, Sattler F, et al. Effects of testosterone supplementation on whole body and regional fat mass and distribution in human immunodeficiency virus-infected men with abdominal obesity. J Clin Endocrinol Metab. 2007;92(3):1049‐1057. [DOI] [PubMed] [Google Scholar]
- 110. Srinivasa S, Fitch KV, Wong K, et al. RAAS activation is associated with visceral adiposity and insulin resistance among HIV-infected patients. J Clin Endocrinol Metab. 2015;100(8):2873‐2882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Flint OP, Noor MA, Hruz PW, et al. The role of protease inhibitors in the pathogenesis of HIV-associated lipodystrophy: cellular mechanisms and clinical implications. Toxicol Pathol. 2009;37(1):65‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Feeney ER, Mallon PW. Impact of mitochondrial toxicity of HIV-1 antiretroviral drugs on lipodystrophy and metabolic dysregulation. Curr Pharm Des. 2010;16(30):3339‐3351. [DOI] [PubMed] [Google Scholar]
- 113. Garrabou G, Lopez S, Moren C, et al. Mitochondrial damage in adipose tissue of untreated HIV-infected patients. AIDS. 2011;25(2):165‐170. [DOI] [PubMed] [Google Scholar]
- 114. Cusi K, Isaacs S, Barb D, et al. American Association of Clinical Endocrinology clinical practice guideline for the diagnosis and management of nonalcoholic fatty liver disease in primary care and endocrinology clinical settings: co-sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr Pract. 2022;28(5):528‐562. [DOI] [PubMed] [Google Scholar]
- 115. Matthews L, Kleiner DE, Chairez C, et al. Pioglitazone for hepatic steatosis in HIV/hepatitis C virus coinfection. AIDS Res Hum Retroviruses. 2015;31(10):961‐966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Sebastiani G, Saeed S, Lebouche B, et al. Vitamin E is an effective treatment for nonalcoholic steatohepatitis in HIV mono-infected patients. AIDS. 2020;34(2):237‐244. [DOI] [PubMed] [Google Scholar]
- 117. Fourman LT, Czerwonka N, Feldpausch MN, et al. Visceral fat reduction with tesamorelin is associated with improved liver enzymes in HIV. AIDS. 2017;31(16):2253‐2259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Stanley TL, Fourman LT, Feldpausch MN, et al. Effects of tesamorelin on non-alcoholic fatty liver disease in HIV: a randomised, double-blind, multicentre trial. Lancet HIV. 2019;6(12):e821‐e830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Erlandson KM, Lake JE. Study overview. Updated 2023-04-13. Accessed January 2023. https://clinicaltrials.gov/study/NCT04216589
- 120. Miljkovic-Gacic I, Gordon CL, Goodpaster BH, et al. Adipose tissue infiltration in skeletal muscle: age patterns and association with diabetes among men of African ancestry. Am J Clin Nutr. 2008;87(6):1590‐1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Delmonico MJ, Harris TB, Visser M, et al. Longitudinal study of muscle strength, quality, and adipose tissue infiltration. Am J Clin Nutr. 2009;90(6):1579‐1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Scherzer R, Shen W, Heymsfield SB, et al. Intermuscular adipose tissue and metabolic associations in HIV infection. Obesity (Silver Spring). 2011;19(2):283‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Adrian S, Miao H, Feng H, et al. Effects of atazanavir, darunavir, and raltegravir on fat and muscle among persons living with HIV. HIV Res Clin Pract. 2020;21(4):91‐98. [DOI] [PubMed] [Google Scholar]
- 124. Alexopoulos N, Katritsis D, Raggi P. Visceral adipose tissue as a source of inflammation and promoter of atherosclerosis. Atherosclerosis. 2014;233(1):104‐112. [DOI] [PubMed] [Google Scholar]
- 125. Lim S, Meigs JB. Links between ectopic fat and vascular disease in humans. Arterioscler Thromb Vasc Biol. 2014;34(9):1820‐1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Gelpi M, Afzal S, Fuchs A, et al. Prior exposure to thymidine analogs and didanosine is associated with long-lasting alterations in adipose tissue distribution and cardiovascular risk factors. AIDS. 2019;33(4):675‐683. [DOI] [PubMed] [Google Scholar]
- 127. Beires MT, Silva-Pinto A, Santos AC, et al. Visceral adipose tissue and carotid intima-media thickness in HIV-infected patients undergoing cART: a prospective cohort study. BMC Infect Dis. 2018;18(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Thiara DK, Liu CY, Raman F, et al. Abnormal myocardial function is related to myocardial steatosis and diffuse myocardial fibrosis in HIV-infected adults. J Infect Dis. 2015;212(10):1544‐1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Holloway CJ, Ntusi N, Suttie J, et al. Comprehensive cardiac magnetic resonance imaging and spectroscopy reveal a high burden of myocardial disease in HIV patients. Circulation. 2013;128(8):814‐822. [DOI] [PubMed] [Google Scholar]
- 130. Toribio M, Neilan TG, Zanni MV. Heart failure among people with HIV: evolving risks, mechanisms, and preventive considerations. Curr HIV/AIDS Rep. 2019;16(5):371‐380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Freiberg MS, Chang CH, Skanderson M, et al. Association between HIV infection and the risk of heart failure with reduced ejection fraction and preserved ejection fraction in the antiretroviral therapy era: results from the veterans aging cohort study. JAMA Cardiol. 2017;2(5):536‐546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Alvi RM, Afshar M, Neilan AM, et al. Heart failure and adverse heart failure outcomes among persons living with HIV in a US tertiary medical center. Am Heart J. 2019;210:39‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Iozzo P. Myocardial, perivascular, and epicardial fat. Diabetes Care. 2011;34(Suppl 2):S371‐S379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. McGavock JM, Victor RG, Unger RH, Szczepaniak LS, American College of Physicians and the American Physiological Society . Adiposity of the heart, revisited. Ann Intern Med. 2006;144(7):517‐524. [DOI] [PubMed] [Google Scholar]
- 135. Neilan TG, Nguyen KL, Zaha VG, et al. Myocardial steatosis among antiretroviral therapy-treated people with human immunodeficiency virus participating in the REPRIEVE trial. J Infect Dis. 2020;222(Suppl 1):S63‐S69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Lo J, Abbara S, Shturman L, et al. Increased prevalence of subclinical coronary atherosclerosis detected by coronary computed tomography angiography in HIV-infected men. AIDS. 2010;24(2):243‐253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Lo J, Abbara S, Rocha-Filho JA, Shturman L, Wei J, Grinspoon SK. Increased epicardial adipose tissue volume in HIV-infected men and relationships to body composition and metabolic parameters. AIDS. 2010;24(13):2127‐2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Buggey J, Yun L, Hung CL, et al. HIV And pericardial fat are associated with abnormal cardiac structure and function among Ugandans. Heart. 2020;106(2):147‐153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Guaraldi G, Scaglioni R, Zona S, et al. Epicardial adipose tissue is an independent marker of cardiovascular risk in HIV-infected patients. AIDS. 2011;25(9):1199‐1205. [DOI] [PubMed] [Google Scholar]
- 140. CDC NDSR . Incidence of newly diagnosed diabetes. Accessed June 2023, 2023. https://www.cdc.gov/diabetes/data/statistics-report/newly-diagnosed-diabetes.html#:∼:text=Incidence%20Among%20Adults, were%20diagnosed%20(Table%202)
- 141. Nansseu JR, Bigna JJ, Kaze AD, Noubiap JJ. Incidence and risk factors for prediabetes and diabetes mellitus among HIV-infected adults on antiretroviral therapy: a systematic review and meta-analysis. Epidemiology. 2018;29(3):431‐441. [DOI] [PubMed] [Google Scholar]
- 142. Wende AR, Symons JD, Abel ED. Mechanisms of lipotoxicity in the cardiovascular system. Curr Hypertens Rep. 2012;14(6):517‐531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Feuerer M, Herrero L, Cipolletta D, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15(8):930‐939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. McDonnell WJ, Koethe JR, Mallal SA, et al. High CD8 T-cell receptor clonality and altered CDR3 properties are associated with elevated isolevuglandins in adipose tissue during diet-induced obesity. Diabetes. 2018;67(11):2361‐2376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15(8):914‐920. [DOI] [PubMed] [Google Scholar]
- 146. Wanjalla CN, McDonnell WJ, Barnett L, et al. Adipose tissue in persons with HIV is enriched for CD4(+) T effector memory and T effector memory RA(+) cells, which show higher CD69 expression and CD57, CX3CR1, GPR56 co-expression with increasing glucose intolerance. Front Immunol. 2019;10:408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Overton ET, Kitch D, Benson CA, et al. Effect of statin therapy in reducing the risk of serious non-AIDS-defining events and nonaccidental death. Clin Infect Dis. 2013;56(10):1471‐1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Rasmussen LD, Kronborg G, Larsen CS, Pedersen C, Gerstoft J, Obel N. Statin therapy and mortality in HIV-infected individuals; a Danish nationwide population-based cohort study. PLoS One. 2013;8(3):e52828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Tsiodras S, Mantzoros C, Hammer S, Samore M. Effects of protease inhibitors on hyperglycemia, hyperlipidemia, and lipodystrophy: a 5-year cohort study. Arch Intern Med. 2000;160(13):2050‐2056. [DOI] [PubMed] [Google Scholar]
- 150. Woerle HJ, Mariuz PR, Meyer C, et al. Mechanisms for the deterioration in glucose tolerance associated with HIV protease inhibitor regimens. Diabetes. 2003;52(4):918‐925. [DOI] [PubMed] [Google Scholar]
- 151. Hertel J, Struthers H, Horj CB, Hruz PW. A structural basis for the acute effects of HIV protease inhibitors on GLUT4 intrinsic activity. J Biol Chem. 2004;279(53):55147‐55152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Noor MA, Flint OP, Maa JF, Parker RA. Effects of atazanavir/ritonavir and lopinavir/ritonavir on glucose uptake and insulin sensitivity: demonstrable differences in vitro and clinically. AIDS. 2006;20(14):1813‐1821. [DOI] [PubMed] [Google Scholar]
- 153. Hresko RC, Hruz PW. HIV Protease inhibitors act as competitive inhibitors of the cytoplasmic glucose binding site of GLUTs with differing affinities for GLUT1 and GLUT4. PLoS One. 2011;6(9):e25237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Noor MA, Parker RA, O’Mara E, et al. The effects of HIV protease inhibitors atazanavir and lopinavir/ritonavir on insulin sensitivity in HIV-seronegative healthy adults. AIDS. 2004;18(16):2137‐2144. [DOI] [PubMed] [Google Scholar]
- 155. Fleischman A, Johnsen S, Systrom DM, et al. Effects of a nucleoside reverse transcriptase inhibitor, stavudine, on glucose disposal and mitochondrial function in muscle of healthy adults. Am J Physiol Endocrinol Metab. 2007;292(6):E1666‐E1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. McLaughlin M, Walsh S, Galvin S. Dolutegravir-induced hyperglycaemia in a patient living with HIV. J Antimicrob Chemother. 2018;73(1):258‐260. [DOI] [PubMed] [Google Scholar]
- 157. Hadigan C, Meigs JB, Rabe J, et al. Increased PAI-1 and tPA antigen levels are reduced with metformin therapy in HIV-infected patients with fat redistribution and insulin resistance. J Clin Endocrinol Metab. 2001;86(2):939‐943. [DOI] [PubMed] [Google Scholar]
- 158. Hsue PY, Waters DD. Time to recognize HIV infection as a major cardiovascular risk factor. Circulation. 2018;138(11):1113‐1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Chow FC, Bacchetti P, Kim AS, Price RW, Hsue PY. Effect of CD4+ cell count and viral suppression on risk of ischemic stroke in HIV infection. AIDS. 2014;28(17):2573‐2577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Lang S, Mary-Krause M, Cotte L, et al. Increased risk of myocardial infarction in HIV-infected patients in France, relative to the general population. AIDS. 2010;24(8):1228‐1230. [DOI] [PubMed] [Google Scholar]
- 161. Feinstein MJ, Hsue PY, Benjamin LA, et al. Characteristics, prevention, and management of cardiovascular disease in people living with HIV: a scientific statement from the American Heart Association. Circulation. 2019;140(2):e98‐e124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Van Hul M, Cani PD. The gut microbiota in obesity and weight management: microbes as friends or foe? Nat Rev Endocrinol. 2023;19(5):258‐271. [DOI] [PubMed] [Google Scholar]
- 163. Dinh DM, Volpe GE, Duffalo C, et al. Intestinal microbiota, microbial translocation, and systemic inflammation in chronic HIV infection. J Infect Dis. 2015;211(1):19‐27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Tuddenham SA, Koay WLA, Zhao N, et al. The impact of human immunodeficiency virus infection on gut microbiota α-diversity: an individual-level meta-analysis. Clin Infect Dis. 2020;70(4):615‐627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365‐1371. [DOI] [PubMed] [Google Scholar]
- 166. Gogokhia L, Taur Y, Juluru K, et al. Intestinal dysbiosis and markers of systemic inflammation in viscerally and generally obese persons living with HIV. J Acquir Immune Defic Syndr. 2020;83(1):81‐89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Felix JB, Cox AR, Hartig SM. Acetyl-CoA and metabolite fluxes regulate white adipose tissue expansion. Trends Endocrinol Metab. 2021;32(5):320‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Dillon SM, Kibbie J, Lee EJ, et al. Low abundance of colonic butyrate-producing bacteria in HIV infection is associated with microbial translocation and immune activation. AIDS. 2017;31(4):511‐521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Herrlich A, Kefaloyianni E, Rose-John S. Mechanisms of interorgan crosstalk in health and disease. FEBS Lett. 2022;596(5):529‐533. [DOI] [PubMed] [Google Scholar]
- 170. Friedman JM. Leptin and the endocrine control of energy balance. Nat Metab. 2019;1(8):754‐764. [DOI] [PubMed] [Google Scholar]
- 171. Ahima RS, Prabakaran D, Mantzoros C, et al. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382(6588):250‐252. [DOI] [PubMed] [Google Scholar]
- 172. Elmquist JK. Hypothalamic pathways underlying the endocrine, autonomic, and behavioral effects of leptin. Physiol Behav. 2001;74(4-5):703‐708. [DOI] [PubMed] [Google Scholar]
- 173. Moon HS, Dalamaga M, Kim SY, et al. Leptin’s role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocr Rev. 2013;34(3):377‐412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Tilişcan C, Mihăilescu R, Munteanu D, et al. Leptin dysfunction and the risk of dyslipidemia and metabolic syndrome in Romanian HIV-infected patients undergoing antiretroviral therapy. BMC Infect Dis. 2014;14(4):O12. [Google Scholar]
- 175. Koethe JR, Moser C, Brown TT, et al. Adipokines, weight gain and metabolic and inflammatory markers after antiretroviral therapy initiation: AIDS Clinical Trials Group (ACTG) A5260s. Clin Infect Dis. 2022;74(5):857‐864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med. 2001;7(8):941‐946. [DOI] [PubMed] [Google Scholar]
- 177. Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 2019;15(9):507‐524. [DOI] [PubMed] [Google Scholar]
- 178. Combs TP, Marliss EB. Adiponectin signaling in the liver. Rev Endocr Metab Disord. 2014;15(2):137‐147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Holland WL, Xia JY, Johnson JA, et al. Inducible overexpression of adiponectin receptors highlight the roles of adiponectin-induced ceramidase signaling in lipid and glucose homeostasis. Mol Metab. 2017;6(3):267‐275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Monczor AN, Li X, Palella FJ Jr, et al. Systemic inflammation characterizes lack of metabolic health in nonobese HIV-infected men. Mediators Inflamm. 2018;2018:5327361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Ketlogetswe KS, Post WS, Li X, et al. Lower adiponectin is associated with subclinical cardiovascular disease among HIV-infected men. AIDS. 2014;28(6):901‐909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Lake JE, Vo QT, Jacobson LP, et al. Adiponectin and interleukin-6, but not adipose tissue, are associated with worse neurocognitive function in HIV-infected men. Antivir Ther. 2015;20(2):235‐244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Samaras K, Gan SK, Peake PW, Carr A, Campbell LV. Proinflammatory markers, insulin sensitivity, and cardiometabolic risk factors in treated HIV infection. Obesity (Silver Spring). 2009;17(1):53‐59. [DOI] [PubMed] [Google Scholar]
- 184. Carr A, Ritzhaupt A, Zhang W, et al. Effects of boosted tipranavir and lopinavir on body composition, insulin sensitivity and adipocytokines in antiretroviral-naive adults. AIDS. 2008;22(17):2313‐2321. [DOI] [PubMed] [Google Scholar]
- 185. Calmy A, Carey D, Mallon PW, et al. Early changes in adipokine levels and baseline limb fat may predict HIV lipoatrophy over 2 years following initiation of antiretroviral therapy. HIV Med. 2008;9(2):101‐110. [DOI] [PubMed] [Google Scholar]
- 186. 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(7):1063‐1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Larsen MA, Isaksen VT, Moen OS, et al. Leptin to adiponectin ratio - a surrogate biomarker for early detection of metabolic disturbances in obesity. Nutr Metab Cardiovasc Dis. 2018;28(11):1114‐1121. [DOI] [PubMed] [Google Scholar]
- 188. Hikasa S, Shimabukuro S, Hideta K, et al. Changes in leptin to adiponectin ratio after antiretroviral therapy: a pilot observational study. Curr HIV Res. Published online March 27, 2023. doi: 10.2174/1570162X21666230327165902 [DOI] [PubMed]
- 189. Cifarelli V, Beeman SC, Smith GI, et al. Decreased adipose tissue oxygenation associates with insulin resistance in individuals with obesity. J Clin Invest. 2020;130(12):6688‐6699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Alessi MC, Peiretti F, Morange P, et al. Production of plasminogen activator inhibitor 1 by human adipose tissue: possible link between visceral fat accumulation and vascular disease. Diabetes. 1997;46(5):860‐867. [DOI] [PubMed] [Google Scholar]
- 191. Fuchs A, Samovski D, Smith GI, et al. Associations among adipose tissue immunology, inflammation, exosomes and insulin sensitivity in people with obesity and nonalcoholic fatty liver disease. Gastroenterology. 2021;161(3):968‐981.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Song C, Burgess S, Eicher JD, O’Donnell CJ, Johnson AD. Causal effect of plasminogen activator inhibitor type 1 on coronary heart disease. J Am Heart Assoc. 2017;6(6):e004918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Ingelsson E, Pencina MJ, Tofler GH, et al. Multimarker approach to evaluate the incidence of the metabolic syndrome and longitudinal changes in metabolic risk factors: the Framingham Offspring Study. Circulation. 2007;116(9):984‐992. [DOI] [PubMed] [Google Scholar]
- 194. Wirunsawanya K, Belyea L, Shikuma C, et al. Plasminogen activator inhibitor-1 predicts negative alterations in whole-body insulin sensitivity in chronic HIV infection. AIDS Res Hum Retroviruses. 2017;33(7):723‐727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Yki-Jarvinen H, Sutinen J, Silveira A, et al. Regulation of plasma PAI-1 concentrations in HAART-associated lipodystrophy during rosiglitazone therapy. Arterioscler Thromb Vasc Biol. 2003;23(4):688‐694. [DOI] [PubMed] [Google Scholar]
- 196. Gruevska A, Moragrega AB, Galindo MJ, Esplugues JV, Blas-Garcia A, Apostolova N. P53 and p53-related mediators PAI-1 and IGFBP-3 are downregulated in peripheral blood mononuclear cells of HIV-patients exposed to non-nucleoside reverse transcriptase inhibitors. Antiviral Res. 2020;178:104784. [DOI] [PubMed] [Google Scholar]
- 197. He G, Andersen O, Haugaard SB, et al. Plasminogen activator inhibitor type 1 (PAI-1) in plasma and adipose tissue in HIV-associated lipodystrophy syndrome. Implications of adipokines. Eur J Clin Invest. 2005;35(9):583‐590. [DOI] [PubMed] [Google Scholar]
- 198. Kharitonenkov A, Shiyanova TL, Koester A, et al. FGF-21 as a novel metabolic regulator. J Clin Invest. 2005;115(6):1627‐1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Markan KR, Naber MC, Ameka MK, et al. Circulating FGF21 is liver derived and enhances glucose uptake during refeeding and overfeeding. Diabetes. 2014;63(12):4057‐4063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Xu J, Lloyd DJ, Hale C, et al. Fibroblast growth factor 21 reverses hepatic steatosis, increases energy expenditure, and improves insulin sensitivity in diet-induced obese mice. Diabetes. 2009;58(1):250‐259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Li H, Gao Z, Zhang J, et al. Sodium butyrate stimulates expression of fibroblast growth factor 21 in liver by inhibition of histone deacetylase 3. Diabetes. 2012;61(4):797‐806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Hotta Y, Nakamura H, Konishi M, et al. Fibroblast growth factor 21 regulates lipolysis in white adipose tissue but is not required for ketogenesis and triglyceride clearance in liver. Endocrinology. 2009;150(10):4625‐4633. [DOI] [PubMed] [Google Scholar]
- 203. BonDurant LD, Ameka M, Naber MC, et al. FGF21 regulates metabolism through adipose-dependent and -independent mechanisms. Cell Metab. 2017;25(4):935‐944.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Fisher FM, Estall JL, Adams AC, et al. Integrated regulation of hepatic metabolism by fibroblast growth factor 21 (FGF21) in vivo. Endocrinology. 2011;152(8):2996‐3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS, Maratos-Flier E. Hepatic fibroblast growth factor 21 is regulated by PPARalpha and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab. 2007;5(6):426‐437. [DOI] [PubMed] [Google Scholar]
- 206. Domingo P, Gallego-Escuredo JM, Domingo JC, et al. Serum FGF21 levels are elevated in association with lipodystrophy, insulin resistance and biomarkers of liver injury in HIV-1-infected patients. AIDS. 2010;24(17):2629‐2637. [DOI] [PubMed] [Google Scholar]
- 207. Payne BA, Price DA, Chinnery PF. Elevated serum fibroblast growth factor 21 levels correlate with immune recovery but not mitochondrial dysfunction in HIV infection. AIDS Res Ther. 2013;10(1):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Srinivasa S, Wong K, Fitch KV, et al. Effects of lifestyle modification and metformin on irisin and FGF21 among HIV-infected subjects with the metabolic syndrome. Clin Endocrinol (Oxf). 2015;82(5):678‐685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Dushay J, Chui PC, Gopalakrishnan GS, et al. Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease. Gastroenterology. 2010;139(2):456‐463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. BonDurant LD, Potthoff MJ. Fibroblast growth factor 21: a versatile regulator of metabolic homeostasis. Annu Rev Nutr. 2018;38(1):173‐196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Flippo KH, Potthoff MJ. Metabolic messengers: FGF21. Nat Metab. 2021;3(3):309‐317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Lee P, Linderman JD, Smith S, et al. Irisin and FGF21 are cold-induced endocrine activators of brown fat function in humans. Cell Metab. 2014;19(2):302‐309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Emmerson PJ, Wang F, Du Y, et al. The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL. Nat Med. 2017;23(10):1215‐1219. [DOI] [PubMed] [Google Scholar]
- 214. Mullican SE, Lin-Schmidt X, Chin CN, et al. GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates. Nat Med. 2017;23(10):1150‐1157. [DOI] [PubMed] [Google Scholar]
- 215. Hsiao EC, Koniaris LG, Zimmers-Koniaris T, Sebald SM, Huynh TV, Lee SJ. Characterization of growth-differentiation factor 15, a transforming growth factor beta superfamily member induced following liver injury. Mol Cell Biol. 2000;20(10):3742‐3751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Coll AP, Chen M, Taskar P, et al. GDF15 mediates the effects of metformin on body weight and energy balance. Nature. 2020;578(7795):444‐448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Paralkar VM, Vail AL, Grasser WA, et al. Cloning and characterization of a novel member of the transforming growth factor-beta/bone morphogenetic protein family. J Biol Chem. 1998;273(22):13760‐13767. [DOI] [PubMed] [Google Scholar]
- 218. Wang D, Day EA, Townsend LK, Djordjevic D, Jorgensen SB, Steinberg GR. GDF15: emerging biology and therapeutic applications for obesity and cardiometabolic disease. Nat Rev Endocrinol. 2021;17(10):592‐607. [DOI] [PubMed] [Google Scholar]
- 219. Keipert S, Ost M. Stress-induced FGF21 and GDF15 in obesity and obesity resistance. Trends Endocrinol Metab. 2021;32(11):904‐915. [DOI] [PubMed] [Google Scholar]
- 220. Patel S, Alvarez-Guaita A, Melvin A, et al. GDF15 provides an endocrine signal of nutritional stress in mice and humans. Cell Metab. 2019;29(3):707‐718.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. O’Neill HM, Maarbjerg SJ, Crane JD, et al. AMP-activated protein kinase (AMPK) beta1beta2 muscle null mice reveal an essential role for AMPK in maintaining mitochondrial content and glucose uptake during exercise. Proc Natl Acad Sci U S A. 2011;108(38):16092‐16097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Moon JS, Goeminne LJE, Kim JT, et al. Growth differentiation factor 15 protects against the aging-mediated systemic inflammatory response in humans and mice. Aging Cell. 2020;19(8):e13195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Adela R, Banerjee SK. GDF-15 as a target and biomarker for diabetes and cardiovascular diseases: a translational prospective. J Diabetes Res. 2015;2015:490842. (Special Issue). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Li D, Zhang H, Zhong Y. Hepatic GDF15 is regulated by CHOP of the unfolded protein response and alleviates NAFLD progression in obese mice. Biochem Biophys Res Commun. 2018;498(3):388‐394. [DOI] [PubMed] [Google Scholar]
- 225. Hung H-C, Wu H-T, Lin C-H, Chou H-W, Ou H-Y, Chang C-J. Associations between GDF15 levels and pre-diabetes in non-obese subjects. J Investig Med. 2022;70(1):79‐84. [DOI] [PubMed] [Google Scholar]
- 226. Agarwal N, Ramirez Bustamante CE, Wu H, et al. Heightened levels of plasma growth differentiation factor 15 in men living with HIV. Physiol Rep. 2022;10(9):e15293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Tsai VW, Zhang HP, Manandhar R, et al. GDF15 mediates adiposity resistance through actions on GFRAL neurons in the hindbrain AP/NTS. Int J Obes (Lond). 2019;43(12):2370‐2380. [DOI] [PubMed] [Google Scholar]
- 228. Isnard S, Lin J, Fombuena B, et al. Repurposing metformin in nondiabetic people with HIV: influence on weight and gut microbiota. Open Forum Infect Dis. 2020;7(9):ofaa338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Bourgi K, Wanjalla C, Koethe JR. Inflammation and metabolic complications in HIV. Curr HIV/AIDS Rep. 2018;15(5):371‐381. [DOI] [PubMed] [Google Scholar]
- 230. Balasubramaniam M, Pandhare J, Dash C. Are microRNAs important players in HIV-1 infection? An update. Viruses. 2018;10(3):110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Mori MA, Thomou T, Boucher J, et al. Altered miRNA processing disrupts brown/white adipocyte determination and associates with lipodystrophy. J Clin Invest. 2014;124(8):3339‐3351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Torriani M, Srinivasa S, Fitch KV, et al. Dysfunctional subcutaneous fat with reduced dicer and brown adipose tissue gene expression in HIV-infected patients. J Clin Endocrinol Metab. 2016;101(3):1225‐1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Thomou T, Mori MA, Dreyfuss JM, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. 2017;542(7642):450‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Srinivasa S, Garcia-Martin R, Torriani M, et al. Altered pattern of circulating miRNAs in HIV lipodystrophy perturbs key adipose differentiation and inflammation pathways. JCI Insight. 2021;6(18):e150399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Hatziioannou T, Evans DT. Animal models for HIV/AIDS research. Nat Rev Microbiol. 2012;10(12):852‐867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Couturier J, Agarwal N, Nehete PN, et al. Infectious SIV resides in adipose tissue and induces metabolic defects in chronically infected rhesus macaques. Retrovirology. 2016;13:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Damouche A, Lazure T, Avettand-Fenoel V, et al. Adipose tissue is a neglected viral reservoir and an inflammatory site during chronic HIV and SIV infection. PLoS Pathog. 2015;11(9):e1005153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Couturier J, Winchester LC, Suliburk JW, et al. Adipocytes impair efficacy of antiretroviral therapy. Antiviral Res. 2018;154:140‐148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Kino T, Gragerov A, Slobodskaya O, Tsopanomichalou M, Chrousos GP, Pavlakis GN. Human immunodeficiency virus type 1 (HIV-1) accessory protein Vpr induces transcription of the HIV-1 and glucocorticoid-responsive promoters by binding directly to p300/CBP coactivators. J Virol. 2002;76(19):9724‐9734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Zou W, Zhang L. Mouse models for studies of in vivo functions of HIV-1 nef. AIDS Rev. 2016;18(3):158‐165. [PubMed] [Google Scholar]
- 241. Agarwal N, Iyer D, Patel SG, et al. HIV-1 Vpr induces adipose dysfunction in vivo through reciprocal effects on PPAR/GR co-regulation. Sci Transl Med. 2013;5(213):213ra164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Fujii Y, Ito M, Ikuta K. Evidence for the role of human immunodeficiency virus type 1 Nef protein as a growth inhibitor to CD4+ T lymphocytes and for the blocking of the Nef function by anti-Nef antibodies. Vaccine. 1993;11(8):837‐847. [DOI] [PubMed] [Google Scholar]
- 243. Agarwal N, Iyer D, Gabbi C, et al. HIV-1 viral protein R (Vpr) induces fatty liver in mice via LXRα and PPARα dysregulation: implications for HIV-specific pathogenesis of NAFLD. Sci Rep. 2017;7(1):13362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Gorwood J, Bourgeois C, Mantecon M, et al. Impact of HIV/simian immunodeficiency virus infection and viral proteins on adipose tissue fibrosis and adipogenesis. AIDS. 2019;33(6):953‐964. [DOI] [PubMed] [Google Scholar]
- 245. Cotter EJ, Chew N, Powderly WG, Doran PP. HIV type 1 alters mesenchymal stem cell differentiation potential and cell phenotype ex vivo. AIDS Res Hum Retroviruses. 2011;27(2):187‐199. [DOI] [PubMed] [Google Scholar]
- 246. Diaz-Delfin J, Domingo P, Wabitsch M, Giralt M, Villarroya F. HIV-1 Tat protein impairs adipogenesis and induces the expression and secretion of proinflammatory cytokines in human SGBS adipocytes. Antivir Ther. 2012;17(3):529‐540. [DOI] [PubMed] [Google Scholar]
- 247. Couturier J, Suliburk JW, Brown JM, et al. Human adipose tissue as a reservoir for memory CD4+ T cells and HIV. AIDS. 2015;29(6):667‐674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Agarwal N, Iyer D, Saha P, et al. HIV-1 viral protein R couples metabolic inflexibility with white adipose tissue thermogenesis. Diabetes. 2021;70(9):2014‐2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Chu VT, Weber T, Graf R, et al. Efficient generation of Rosa26 knock-in mice using CRISPR/Cas9 in C57BL/6 zygotes. BMC Biotechnol. 2016;16(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Kim KH, Hartig SM. Contributions of microRNAs to peripheral insulin sensitivity. Endocrinology. 2022;163(2):bqab250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Arama V, Tiliscan C, Streinu-Cercel A, et al. Insulin resistance and adipokines serum levels in a Caucasian cohort of HIV-positive patients undergoing antiretroviral therapy: a cross sectional study. BMC Endocr Disord. 2013;13(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Long MT, Pedley A, Massaro JM, et al. A simple clinical model predicts incident hepatic steatosis in a community-based cohort: the Framingham Heart Study. Liver Int. 2018;38(8):1495‐1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Das B, Dobrowolski C, Luttge B, et al. Estrogen receptor-1 is a key regulator of HIV-1 latency that imparts gender-specific restrictions on the latent reservoir. Proc Natl Acad Sci U S A. 2018;115(33):E7795‐E7804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Maurice JB, Patel A, Scott AJ, Patel K, Thursz M, Lemoine M. Prevalence and risk factors of nonalcoholic fatty liver disease in HIV-monoinfection. AIDS. 2017;31(11):1621‐1632. [DOI] [PubMed] [Google Scholar]
- 255. Erlandson KM, Zhang L, Lake JE, et al. Changes in weight and weight distribution across the lifespan among HIV-infected and -uninfected men and women. Medicine (Baltimore). 2016;95(46):e5399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Soti S, Corey KE, Lake JE, Erlandson KM. NAFLD and HIV: do sex, race, and ethnicity explain HIV-related risk? Curr HIV/AIDS Rep. 2018;15(3):212‐222. [DOI] [PMC free article] [PubMed] [Google Scholar]



