Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
Next Article in Journal
Advances in Leishmania Vaccines: Current Development and Future Prospects
Previous Article in Journal
Review of the Highly Pathogenic Avian Influenza in Argentina in 2023: Chronicle of Its Emergence and Control in Poultry
Previous Article in Special Issue
Testing Hepatitis E Seroprevalence among HIV-Infected Patients in Greece: The SHIP Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bone Loss and Fractures in Post-Menopausal Women Living with HIV: A Narrative Review

by
Maryam Jamshaid
1,2,
Amirmohammad Heidari
1,2,
Ahmed Hassan
3,
Dushyant Mital
4,
Oliver Pearce
5,
Maria Panourgia
6,7 and
Mohamed H. Ahmed
6,7,8,*
1
Department of Trauma and Orthopaedics, Liverpool University Hospital NHS Trust, Liverpool L69 3BX, UK
2
School of Medicine, University of Liverpool, Liverpool L69 3BX, UK
3
Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
4
Department of HIV and Blood Borne Virus, Milton Keynes University Hospital NHS Foundation Trust, Eaglestone, Milton Keynes MK6 5LD, UK
5
Department of Trauma and Orthopaedics, Milton Keynes University Hospital NHS Foundation Trust, Milton Keynes MK6 5LD, UK
6
Department of Geriatric Medicine, Milton Keynes University Hospital NHS Foundation Trust, Eaglestone, Milton Keynes MK6 5LD, UK
7
Faculty of Medicine and Health Sciences, University of Buckingham, Buckingham MK18 1EG, UK
8
Department of Medicine and HIV Metabolic Clinic, Milton Keynes University Hospital NHS Foundation Trust, Eaglestone, Milton Keynes MK6 5LD, UK
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(9), 811; https://doi.org/10.3390/pathogens13090811
Submission received: 18 August 2024 / Revised: 7 September 2024 / Accepted: 11 September 2024 / Published: 19 September 2024

Abstract

:
Introduction: Post-menopausal women living with Human Immunodeficiency Virus (WLHIV) face an increased risk of bone fractures due to the relationship between HIV-related factors and menopause. This narrative review aims to summarise the current knowledge about fracture risk among post-menopausal WLHIV in particular looking at hormonal changes, combined antiretroviral therapy (cART), lifestyle factors, and psychosocial implications. We also profiled a summary of the significant, recent studies of post-menopausal WLHIV residing in low-income countries (LIC). Methods: A thorough search of the literature was performed across PubMed, Medline, Scopus, and Google Scholar, focussing on studies published between 2000 and 2024. Inclusion criteria entailed original research, reviews, and meta-analyses addressing bone mineral density (BMD), fracture incidence, and related risk factors in post-menopausal WLHIV. Results: The review identified 223 relevant studies. Post-menopausal WLHIV exhibit significantly lower BMD and higher fracture rates compared to both HIV-negative post-menopausal women and pre-menopausal WLHIV. cART, particularly tenofovir disoproxil fumarate (TDF), contributes to reduced BMD. Menopausal status exacerbates this risk through decreased oestrogen levels, leading to increased bone resorption. Moreover, lifestyle choices such as smoking, alcohol consumption, and low physical activity are more prevalent in PWHIV, which further elevates fracture risk. Different psychosocial factors may make WLWHIV more vulnerable at this stage of their life, such as depression, isolation, stigma, and housing and nutritional issues. Women living in LICs face a variety of challenges in accessing HIV care. There are gaps in research related to the prevalence of osteoporosis and bone loss in post-menopausal WLHIV in LICs. Conclusion: Post-menopausal women living with HIV face a significantly higher risk of bone loss and fractures due to the combined effects of HIV and menopause. Antiretroviral therapy (particularly TDF), lifestyle factors, and psychosocial challenges exacerbate this risk. There is a need for careful selection of cART, hormone replacement therapy (HRT), and emerging treatments such as Abaloparatide. A holistic approach including lifestyle changes and psychosocial support is crucial to reduce fracture risk in WLHIV, especially in low-income countries.

1. Introduction

Over the past 25 years, combination antiretroviral therapy (cART) has transformed HIV care from a terminal disease into a chronic condition with normal life expectancy and stability for those who have access to treatment [1]. Before the introduction of combination antiretroviral therapy (cART) in the mid-1990s, HIV was considered a terminal disease. Typically, the virus would progress to AIDS within 10–12 years, and once AIDS was diagnosed, life expectancy dropped to 1–2 years due to opportunistic infections and immune system failure [2]. However, the advent of cART transformed HIV into a manageable chronic condition. For individuals who start cART early, particularly with a CD4 count above 350 cells/mm3, life expectancy can be similar to that of the general population. For example, in the UK, a 20-year-old starting cART with a CD4 count between 200–350 cells/mm3 has a life expectancy of around 53.4 years, close to the general population’s figures [3].
In addition to the remarkable improvements in life expectancy and mortality, the epidemiology of HIV has evolved to affect females more significantly [4]. In 2023, women accounted for more than half of 39.9 million people living with HIV (PLHIV) globally [5]. In combination with the ageing population increasing globally, the percentage of women over 50 living with HIV is also increasing [6]. Between 2000 and 2024, the trend of HIV affecting women has seen significant shifts [7]. In 2000, women accounted for a lower proportion of global HIV infections, but over time, their share has increased, particularly among young women and adolescent girls aged 15–24 [7]. By 2023, females represented 44% of new global infections, with adolescent girls being four times more likely than boys to contract HIV in this age group. Despite a 63% reduction in new infections among adolescent girls and young women since 2000, progress has slowed in recent years, with 210,000 new cases reported in 2023, compared to a 2025 target of 50,000 [7,8].
Biological menopause is a normal part of ageing for women, and this transition has historically been linked to detrimental consequences in women in terms of bone loss [9]. The World Health Organization (WHO) defines biological menopause as the permanent cessation of menses (also known as amenorrhoea) for a period of at least twelve months [10]. While the age of onset of menopause varies with individual factors and geographical location, it typically occurs between the ages of 45–56 for women without HIV [11]. This period of amenorrhoea is caused by non-pathological oestrogen deficiency and a resultant loss of follicular activity in the ovaries, which can increase the risk of bone loss [11]. HIV has been shown to lead to a significantly higher risk of hip and major osteoporotic fractures [12], and thus women living with HIV (WLHIV) experience an intricate, bidirectional relationship between menopause and HIV, which further adds an additional layer of complexity to the natural ageing process.
WLHIV have also been shown to experience premature menopause. This population is more likely to experience early (before age 45) or premature menopause (before age 40) compared to HIV-negative women. The median age of menopause in HIV-positive women is around 48 years, with early menopause rates ranging from 14.6% to 27.9% and premature menopause rates between 2.3% and 35% [13].
Research on the impact of HIV on ovarian ageing has revealed multiple mechanisms that contribute to early ovarian failure in women living with HIV. Several studies have focused on anti-Müllerian hormone (AMH), a key marker of ovarian reserve, finding that women with HIV generally have lower AMH levels compared to HIV-negative women [14,15]. This suggests a depletion of ovarian reserve, which could indicate accelerated ovarian aging. Lower AMH levels have been linked to chronic inflammation caused by HIV infection. Even with the use of antiretroviral therapy (ART), this inflammation persists [16]. Chronic immune activation, driven by low-level viral replication, microbial translocation from the gut, and ART toxicity, increases the levels of pro-inflammatory cytokines such as IL-1, IL-6, and TNF-α, which damage ovarian follicles and contribute to premature ovarian insufficiency [14,17]. These inflammatory processes and persistent immune activation seem to play a central role in driving early ovarian failure in women with HIV [16].
Moreover, studies have shown that mitochondrial dysfunction, induced by both HIV and long-term ART, accelerates ageing processes within the ovaries [18]. Mitochondrial damage leads to cellular senescence, oxidative stress, and apoptosis, all of which are detrimental to ovarian health [19]. Certain ART medications, such as non-nucleoside reverse transcriptase inhibitors (NNRTIs) and protease inhibitors (PIs), are known to exacerbate this mitochondrial dysfunction, leading to further reproductive ageing [18]. Additionally, as women with HIV transition into menopause, the depletion of oestrogen aggravates immune dysregulation, further increasing the viral reservoirs and immune activation [17].
These hormonal changes are linked to increased inflammation and microbial translocation, which worsen immune responses and lead to further reproductive ageing [19]. Overall, these findings highlight a complex interaction between chronic inflammation, ART-induced toxicity, mitochondrial dysfunction, and hormonal changes, which accelerate reproductive aging and ovarian failure in women with HIV.
In a recent review article, Ahmed et al. concluded that menopause seems to occur at least four to five years earlier in HIV-infected women compared to the general population [20]. Moreover, Chikwati et al. showed that age at menopause in sub-Saharan Africa was lower in WLHIV than in HIV-negative women (48.1 ± 5.1 vs. 50.9 ± 4.7 years, p < 0.001) [21]. They also noted that menopause can be associated with metabolic changes such as insulin resistance, dyslipidaemia, and the presence of atherosclerosis.
A complex clinical process, menopause is a unique experience for each woman, which leads to a wide range of changes in endocrine and other age-related co-morbidities in addition to changes in bone health [22]. HIV has particularly been shown to be associated with evidence of osteoporosis across all menopause stages, unlike those without HIV [23]. In post-menopausal females, the WHO defines osteoporosis as bone mineral density (BMD) measurement taken using dual X-ray absorptiometry (DEXA) at the spine, hip, or forearm that should show a result that is at least 2.5 standard deviations below that of a “young normal” adult (T-score ≤ −2.5), or there should be a history of one or more fragility fractures [24]. While primary osteoporosis can be a normal part of ageing and menopause, secondary osteoporosis is bone loss due to clinical disorders such as endocrine pathology, which often occurs in HIV-infected individuals [25]. Up to 30% of post-menopausal women have secondary osteoporosis [26] and any resultant bone fractures can have devastating consequences on the individual as they can lead to prolonged and costly hospital admissions. Globally, the overall mean one-year mortality rate following a hip fracture is 22.0% [27], which may undermine the improvements in quality of life achieved through cART.
Most research on co-morbidities in PLWHIV has focused on the male population or a combination of both males and females. There has been limited research that specifically addresses bone loss, fractures, and related co-morbidities experienced specifically by WLHIV. Importantly, the concept of effective menopausal care (which constitutes a continuum of symptom management and optimisation of medical health, including cardiovascular, bone, and mental health) is rarely implemented in WLHIV. Dragovic et al., in a recent review on menopause care in the UK, revealed that few HIV specialists are fully trained in menopausal medicine, and general practitioners, or primary care physicians, are cautious when treating menopausal symptoms in WLHIV due to drug-drug interactions with cART and hormone replacement therapies (HRT) [28]. They emphasised that WLHIV experience significant distress due to having to shuffle between specialists to meet their menopausal and HIV needs [28]. Zahn et al. showed there are few interventions that exist to address disparities in menopausal care, even for HIV-negative women [29]. WLHIV thus experience a compounded lack of high-quality, individualised menopause care, which would lead to improved patient experience. It is important to highlight that a diagnosis of menopause with HIV is still dependent on HIV physician judgement due to a lack of consensus. For instance, Scofield et al. demonstrated disparities among HIV guidelines in the diagnosis of menopause and the assessment and treatment of menopausal symptoms [30]. They included five HIV and six general menopause management guidelines published between 2015 and 2023, and five menopausal symptom assessment scales. They found that the HIV guidelines did not offer guidance on how to diagnose menopause or how to differentiate between HIV-related and menopause-related symptoms. Importantly, the scales for menopausal symptom assessment were not validated specifically for WLHIV. The authors recommended the need for the development of a comprehensive guideline that addresses all relevant factors in managing menopause in women with HIV [30].
The aim of this narrative review is to elucidate the effects of bone loss and related co-morbidities of HIV in post-menopausal women, including those living in LICs, and recommend key management areas by drawing on existing research to highlight key findings, mechanisms, and clinical implications. We much hope this review will contribute to establishing an initiative to promote the concept of effective menopausal care among women living with HIV.

2. Methods

This narrative review article was conducted by undertaking a comprehensive search across PubMed, Medline, Scopus, and Google Scholar, focussing on studies published between 2000 and 2023. Keywords included “Osteoporosis” OR “Bone density*” OR “Bone loss*”, “Postmenopausal*” OR “Menopause*”, “Risk factor*” OR “Prevent*” OR “Treat*” OR “Calcium” OR “Vitamin D” OR “Frac*”, “HIV” OR “Human immunodeficiency virus” AND “ageing woman” OR “post-menopause” OR “post-menopausal”. Inclusion criteria entailed original research, reviews, and meta-analyses addressing BMD, fracture incidence, and related risk factors in post-menopausal WLHIV. Only articles published in the English language were included. Figure 1 shows how the literature search was conducted and the number of studies included in this review article. In this narrative review, we evaluated a broad range of studies to understand bone loss and fracture risk in post-menopausal women living with HIV. The inclusion and exclusion criteria used in this review are listed below.

2.1. Inclusion Criteria

We included studies published between 2000 to 2024.
Study types included: epidemiological studies, clinical trials, systematic reviews and meta-analyses, observational studies, pathophysiological studies, animal studies, and in vitro studies.
Population focus: the primary population was post-menopausal WLHIV, with studies from various geographical regions. The review criteria included papers for comparisons with pre-menopausal WLHIV to highlight the impact of menopause, with HIV-negative post-menopausal women to isolate the effects of HIV and ART on bone health, and with men living with HIV to contextualise gender differences in bone health outcomes.
Relevant studies had the following outcomes of interest:
  • Bone Mineral Density (BMD)
  • Fracture Incidence and Risk
  • Bone Turnover Markers
  • Vitamin D Status
  • Impact of Antiretroviral Therapy (ART)
  • FRAX Score and Fracture Risk Prediction
  • Influence of Co-Morbidities and Lifestyle Factors

2.2. Exclusion Criteria

1.
Population-specific exclusions:
Studies not involving women. (Include both pre- and post-menopausal women, but studies focussing only on men are excluded.)
Studies not including HIV-infected individuals.
2.
Outcome-specific exclusions:
Studies that do not address bone health (such as BMD, fractures, or osteoporosis).
Research that focusses on unrelated health outcomes (e.g., cardiovascular disease, mental health) without mentioning bone health or fracture risk.
3.
Study design exclusions:
Non-peer-reviewed articles (e.g., conference abstracts, editorials, letters).
Studies not based on human subjects (exclude animal or laboratory studies).
Case reports or small case series that do not provide broad or generalisable findings.
4.
Geographical or setting exclusions:
Studies conducted in regions not relevant to the global HIV burden, such as those focused solely on populations where HIV and osteoporosis are not common comorbidities.
5.
Language exclusions:
Studies not published in English.
6.
Publication date exclusions:
Studies published before 2000, to focus on more recent data relevant to current HIV treatments and management of bone health.
To ensure the robustness of our findings, we prioritised higher-quality evidence such as cohort studies, systematic reviews, and meta-analyses. Cross-sectional studies and case reports were included for additional context but recognised as less rigorous. Only studies directly assessing relevant outcomes, such as bone mineral density (BMD) and fracture incidence, were considered, with preference given to those using validated tools such as dual-energy X-ray absorptiometry (DXA) scans. We also prioritised studies that controlled for key confounding factors such as age, body mass index (BMI), menopausal status, and HIV treatment regimen to minimise bias. Geographical and temporal relevance were also key considerations. We placed an emphasis on studies conducted in low-income countries, where the burden of HIV is particularly high, and limited our scope to studies published between 2000 and 2023 to ensure relevance to modern treatment approaches. While we did not formally apply a bias assessment tool, each study was critically appraised for methodological soundness, with particular attention to potential sources of bias, such as selection and reporting biases. This approach ensured that our review provides a comprehensive yet methodologically sound overview of the current evidence.

3. Results

3.1. Epidemiology

In 2023, women made up 53% of the total 38.4 million PLWHIV [2]. The proportion of women affected by HIV varies by geographical location. Although sub-Saharan Africa (SSA) accounts for 12% of the world population, the region contains 70% of all PLWHIV [31]. In SSA, women and girls account for 63% of all HIV diagnoses [2]. In the UK, women accounted for one-third of all PLWHIV in 2022 [32]. The United Nations Population Fund (UNFPA) has revealed that the global share of people aged 65 and over has nearly doubled from 5.5% in 1974 to 10.3% in 2024 [33]. Due to the increasing ageing population, it is estimated that by 2030, nearly 70% of all PLWHIV will be above the age of 50 in the developed world, thus increasing the proportion of WLHIV who are post-menopausal [34].
This substantial rise in the number of post-menopausal women accessing care in the UK is reflected in the PRIME (Positive Transitions Through the Menopause) study [35]. One of the largest studies globally, the PRIME study approached 1999 women in the UK between June 2015 and April 2018 [17]. It revealed that 10,350 WLHIV, aged between 45 and 56 (and therefore, possibly menopausal), accessed HIV care in 2016, which is a five-fold increase from the number of women in this age range accessing HIV care in 2006 and half the total number of WLHIV who attended HIV care [17].
Menopause-associated rapid bone loss leads to a two- to three-fold greater fracture incidence in women [36]. Almost 50% of women above the age of 50 will experience a fracture [37]. A recent systematic review has demonstrated a varying prevalence of osteoporosis in post-menopausal women, ranging from 7% to 84% in WLHIV and 0.7% to 23% in HIV-negative women [38]. This wide range reflects differences in study populations, ART exposure, lifestyle factors, and regional disparities. The highest prevalence of osteoporosis (84%) in WLHIV is approximately 3.65 times higher than the maximum prevalence observed in HIV-negative women (23%). This significant difference likely stems from HIV-related chronic inflammation, ART exposure, and other contributing factors that exacerbate bone loss in WLHIV, particularly during menopause. The review also highlighted that post-menopausal WLHIV report a fracture prevalence 1.5 times greater than their non-HIV infected counterparts [38]. Moreover, results from the Women’s Interagency HIV Study showed that HIV infection and menopausal stage were independent predictors of lower BMD and had an additive effect on the lumbar spine and total hip BMD [39]. Similarly, Leite-Silva et al. showed that WLHIV in Brazil are more likely to have osteopenia/osteoporosis and a high FRAX index score [40].
To reconcile the variations between these studies, the data highlight that regional factors, ART regimen, and lifestyle differences contribute to the variability in osteoporosis prevalence and fracture risk in WLHIV. The findings from the Women’s Interagency HIV Study suggest that HIV’s impact on bone mineral density may vary across populations, and further research is needed to explore these discrepancies. More research is needed to fully understand the additive effects of HIV and menopause on bone health across diverse WLHIV populations [41]. Table 1 summarises several studies examining the risk of fractures in post-menopausal women living with HIV (WLHIV). The findings show that WLHIV have a higher risk of fractures, particularly in the lumbar spine, femoral neck, and hips, compared to non-HIV-infected women. One study found that WLHIV had a 6.3% greater chance of vertebral fractures and lower bone mineral density (BMD) at the femoral neck and hip, with 22 out of 104 participants diagnosed with osteoporosis but not receiving treatment. Another study revealed that older HIV-positive women were 2.11 times more likely to suffer hip fractures compared to non-HIV women. Additionally, a Brazilian study indicated that WLHIV are more prone to osteoporosis and have a higher FRAX score, which estimates the risk of major osteoporotic fractures. Overall, Table 1 highlights the increased fracture risk in WLHIV, emphasising the importance of monitoring bone health in this population to prevent fractures.
It is important to note that PLWHIV experience greater postoperative complications following surgical treatment for the above examples of fractures. Anand et al. showed that due to decreasing CD4+ counts, PLWHIV experience poorer immunity and are more likely to contract opportunistic infections in the postoperative period [46]. Furthermore, in a retrospective study of 37 patients, Akkaya et al. demonstrated that the in-hospital complication rate for PLWHIV following total knee arthroplasty (TKA) or total hip arthroplasty (THA) was 10.8% (complications included delirium, acute renal failure, deep vein thrombosis, and trochanteric fracture) [47]. In contrast, a recent systematic review revealed a systemic medical complication rate of 5.1% following THA and 6.9% following TKA in non-HIV-infected patients [48].

3.2. Pathophysiology

In this section, we will discuss normal bone loss and the pathophysiology of HIV and its direct and indirect effects on systems that lead to bone loss. The pathophysiology of HIV directly impacts the immune system and indirectly impacts reproductive hormones, metabolic disease, and thyroid hormones. We will then discuss how patients with HIV are often concurrently found to have nutritional deficiencies and co-infection with Hepatitis C, which also contributes to bone loss, especially in post-menopausal WLHIV.

3.2.1. Normal Bone Loss Due to the Ageing Process

Following peak bone mineral density in early adulthood (25–35 years of age), the skeleton undergoes constant bone remodelling in both men and women [49]. Skeletal remodelling is homeostatic, with osteoclasts (causing bone resorption) and osteoblasts (synthesising new bone cells) in harmony with each other [50]. Giant multinucleated cells of hematopoietic (myeloid) origin, osteoclasts resorb bone under the coordinated function of two key cytokines, the receptor activator of NF-κB (RANKL) and osteoprotegerin (OPG) [51]. RANKL is a key cytokine that drives osteoclast differentiation and subsequent activity. OPG acts as a decoy receptor for RANKL; thereby, it moderates bone resorption [52]. Therefore, the rate of bone resorption in the body is determined by the ratio of RANKL and OPG [52]. Normal ageing leads to an increase in OPG by B-cells to target excessive RANKL activity, which, if left unchecked, may lead to high rates of bone resorption [53]. Despite the increase in OPG, ageing, overall, leads to a gradual loss of BMD as the rate of new bone formation drops below the rate of resorption [49]. Over four to eight years, post-menopausal women experience a 20–30% loss of trabecular bone and 5–10% loss of compact bone [51]. This is crucial, as a remarkable number of WLHIV experience early menopause. Importantly, direct factors such as the HIV virus and menopause will have a substantial impact on increasing bone loss in the majority of WLHIV. In terms of indirect factors (diabetes, thyroid disorders, fatty liver, and hepatitis), even though they are commonly associated with HIV, not all WLHIV will encounter them. In addition, HIV medication per se is also known to be associated with bone loss. In the following sections, we attempt to provide a summary of all these factors. A brief summary of the direct and indirect effects of HIV on reduced BMD is demonstrated in Figure 2 below.
Direct impact of HIV on bone loss. HIV per se as a cause of ongoing chronic inflammation: PLWHIV have historically been known to demonstrate lower BMD [49]. A pooled odds ratio of 3.7 has been demonstrated by a recent meta-analysis that compared BMD in HIV-infected versus non-HIV-infected persons, thereby highlighting the accelerated rate of bone loss in PLWHIV [10]. Another study showed that people living with HIV (PLWHIV) have a higher risk of reduced BMD and fractures due to HIV-induced chronic inflammation and the effects of antiretroviral therapy (ART) [54]. Similarly, a review showed that PLWHIV experience accelerated bone loss, with additional contributions from lifestyle and nutritional factors. These studies collectively highlight the need for focused bone health monitoring in PLWHIV [55].
The exact causes and mechanisms of bone loss directly due to the virological nature of HIV remain unknown. However, risk factors contributing to accelerated bone loss have been demonstrated in HIV-infected communities, including social factors such as smoking and excessive alcohol, as well as HIV-related effects such as low BMI, hypogonadism, and kidney disease [56]. PLWH is a particularly vulnerable cohort prone to problematic health behaviours and are more likely to drink alcohol and use drugs [57]. The immune-skeletal interface (ISI) is a complex system of shared cells and cytokines through which the immune system directly impacts the skeletal system [58]. In the context of HIV infection, the chronic inflammation diminishes the compensatory increase in OPG that normally occurs during ageing, thereby leading to increased RANKL production. This may lead to an exacerbated loss of bone [58]. HIV-infected T-cells can modulate the function of macrophages and lymphocytes and, most importantly, enhance the recruitment of osteoclasts, leading to an increase in RANKL and low expression of OPG [59]. Recent research into the molecular pathways has revealed that HIV-infected T-cells and B-cells, specifically those with prolonged viral exposure, cause persistent upregulation of RANKL, particularly through the production of soluble RANKL by activated T-cells. This soluble form of RANKL plays a more significant role than previously understood in directly stimulating osteoclast precursor cells, contributing to sustained osteoclastogenesis in PLWHIV [60,61]. A five-fold increase in RANKL production from B-cells was observed in HIV patients compared to non-HIV controls, leading to amplified osteoclast differentiation and bone resorption. Furthermore, the reduction in OPG in these individuals, exacerbated by chronic HIV-driven immune dysregulation, significantly contributes to this imbalance, favouring excessive bone breakdown [62].
Unfortunately, additional cellular processes continue to erode the bone; for example, the osteoclast in HIV activates RANKL, tartrate-resistant acidic phosphatase (TRAP), and cathepsin K, and this leads to more osteoclast activity, thus decreasing bone density [63]. Molecular studies have shown that TRAP and cathepsin K expression is significantly upregulated in PLWHIV, further intensifying osteoclast-mediated bone matrix degradation. The upregulation of these key enzymes results in more aggressive bone resorption, contributing to a heightened loss of bone mineral density and overall skeletal fragility [64,65]. Besides decreasing the osteoblasts or osteocalcin, HIV modulates the bone marrow mesenchymal stem cells, and this leads to a release of interleukin-6 (IL-6), interleukin 8 (IL-8), alkaline phosphatase (ALP), runt-related transcription factor 2 (RUNX-2), and bone morphogenic proteins (BMP-2, BMP-7) [66]. Chronic inflammation plays a central role in HIV-induced bone loss. Elevated levels of pro-inflammatory cytokines, especially IL-6 and TNF-α, have been identified as major contributors to increased bone resorption. IL-6, which is upregulated in PLWHIV, directly correlates with reduced BMD and stimulates the differentiation and activation of osteoclasts by increasing RANKL expression. IL-6 also disrupts osteoblast activity, inhibiting bone formation, thus further exacerbating bone loss [60].
All of these factors work toward decreasing bone formation and increasing osteoclastic activities (bone resorption) [67] Studies on HIV-transgenic rats, an animal model of HIV infection that lacks many of the confounding influences on bone that are associated with human HIV patients revealed a significant 75% decline in expression of bone marrow OPG due to defective B cell OPG production [67] This loss of OPG, coupled with a dramatic five-fold increase in receptor RANKL production by B-cells, accounted for a significant increase in osteoclast differentiation [67]. Overall, In vivo markers of bone resorption increased by 43%, while bone formation markers remained unchanged. This increase in the rate of bone resorption led to a significant loss of BMD, which was demonstrated using DXA, with a percent change between HIV transgenic rats and control groups of −36% in femurs, −29% in tibias and −21% in the lumbar spine. Moreover, cortical volume of bones was also significantly reduced by −18.7% [67]. In the context of women and menopause, both B-cells and T-cells have been shown to be key sources of enhanced RANKL production in post-menopausal women relative to premenopausal women and post-menopausal women receiving oestrogen replacement [68].
Overall, as described above, the highlighted cascade of events illustrates how HIV worsens bone health by enhancing bone resorption. HIV stimulates mesenchymal stem cells to release cytokines (IL-6, IL-8), ALP, and BMPs, promoting osteoclast activity while reducing bone formation. In HIV-transgenic rats, a 75% reduction in OPG and a five-fold increase in RANKL significantly increased osteoclast differentiation, leading to a marked decrease in BMD (36% in femurs, 29% in tibias). This is critical for PLWHIV, especially post-menopausal women, as it accelerates bone loss and increases fracture risk. (Figure 2)
Gonadal hormones and effects on age at menopause. Among HIV-infected women, early menopause is frequent [20]. Serum plasma levels of anti-Mullerian hormone (AMH), an indicator of ovarian reserve and FSH, are highly predictive of the age onset of menopause [69]. In WLHIV, AMH has been documented to be significantly lower, directly associated with the early onset of menopause [69,70]. Another study found that AMH is a strong predictor of the timing of the final menstrual period in HIV-infected women [71]. However, other factors can additionally cause early menopause, such as low BMI, ethnicity, smoking, low CD4+ cell count, hepatitis C virus coinfection, and a history of Acquired Immunodeficiency Syndrome (AIDS) [20,72] The mechanism of this correlation is multivariant, as one of the most prevalent predictors of early onset of menopause is the use of tobacco inhalation inhalation [51]. Smoking is common among WLHIV and is often accompanied with the use of recreational drugs [73]. The use of cocaine and heroin five years before the onset of menopause is associated with early-onset menopause [74].
A recent survey of 102 WLHIV showed that the age of menopause in WLHIV ranged between 39 and 49 years (median age: 46), while it was between 44.5 and 48 years (median age: 47) in uninfected women [74].
Ethnicity, in particular, has also been shown to play a role in early menopause in WLHIV. A retrospective Swiss study demonstrated that Black women were found to be four times more likely to experience early menopause than women of other ethnicities (aOR = 4.2, 95% CI: 2.5–7.2, p < 0.001) [72]. Interestingly, traditional risk factors such as smoking, drug use, and body weight did not significantly impact the age of menopause, indicating that ethnicity plays a dominant role [72]. HIV-related factors such as low CD4 counts, viral suppression, or hepatitis coinfection also showed no effect. Being underweight was initially associated with a later menopause but was excluded due to confounding with Black ethnicity. Despite Black women being more likely to be overweight (70% vs. 32%) and less likely to smoke (10% vs. 57%) or use drugs (1% vs. 20%), they still experienced earlier menopause [72]. This highlights that biological factors and possibly socioeconomic disparities are stronger determinants of early menopause in this population than lifestyle factors [72].
Additionally, a study in Burkina Faso with 93 patients revealed that early menopause occurred in 5.5% of WLHIV but only occurred in 1.4% of a non-infected African population [75].
Indirect impact of HIV on metabolic disorders and bone. Diabetes, dyslipidaemia, and obesity. The menopause transition leads to increased risk of cardiovascular disease (CVD) due to factors such as the loss of the protective effects of oestrogen. Lipid profile changes that lead to atherogenesis, such as increased low-density lipoprotein (LDL), total cholesterol, and apoprotein B, have been noted in peri-menopausal and post-menopausal women [76]. These factors have been independently associated with abdominal fat, age, and menopause [76]. Reduced HDL levels and increased insulin resistance due to changes in androgen levels have been exhibited during menopause [77]. This leads to an increased risk of developing type II diabetes, non-alcoholic fatty liver disease (NAFLD), metabolic associated fatty liver disease (MFFLD), and metabolic syndrome [78].
For individuals with HIV who are not on ART, there is a decrease in total cholesterol (TC), LDL cholesterol (LDL-C), and HDL cholesterol (HDL-C) compared to pre-seroconversion levels, while triglycerides (TG) show no significant change compared to individuals without HIV [79,80]. When people living with HIV begin ART, TC and LDL-C levels tend to increase compared to pre-ART levels. HDL-C generally shows no change, and the effect on TG levels varies depending on the type of ART being used [80,81]. In cases of advanced HIV or AIDS, lipid levels behave differently. There is a decrease in TC, LDL-C, and HDL-C compared to individuals without HIV, while TG levels tend to increase, particularly when compared to both individuals without HIV and those with HIV but without AIDS [79,80]. Lipodystrophy, particularly in patients undergoing ART, contributes significantly to these metabolic disturbances. The redistribution of fat (e.g., loss of subcutaneous fat and increased visceral adiposity) promotes a chronic inflammatory state, marked by increased TNF-α and IL-6, which not only worsens insulin resistance but also disrupts bone health by increasing bone resorption and reducing bone formation. This process elevates the risk of osteopenia and osteoporosis in PLWHIV [82,83,84].
PLHIV are at a higher risk of CVD, hypertension, and diabetes due to multiple factors, such as specific combination antiretroviral therapy (cART) medications, inflammatory effects of HIV, and traditional risk factors such as dyslipidaemia [85,86]. Additionally, risk factors such as smoking and poor fitness levels, which increase the risk of CVD and associated metabolic disorders, are more common in PLWHIV [87,88]. PLWHIV are known to be in a hypermetabolic state with an elevated resting energy expenditure mediated by IL-1, TNF, and interferon-α.
Insulin resistance, commonly associated with HIV-related lipodystrophy, further exacerbates bone loss. The reduced insulin sensitivity impairs the anabolic effects of insulin on bones, leading to increased osteoclast activity and a weakening of bone integrity. This is particularly concerning in PLWHIV, who often experience a combination of chronic inflammation, visceral fat accumulation, and dysregulated glucose metabolism, contributing to accelerated bone degradation [89,90,91].
Normal growth hormone (GH) levels are essential for maintaining protein balance and muscle mass. Patients with advanced HIV- and AIDS-related wasting type syndromes routinely have elevated GH and low insulin-like growth factor (IGF)-1 levels, suggestive of GH resistance. Testosterone levels also correlate closely with lean muscle mass as WLHIV tend to display lower testosterone levels, leading to a loss of muscle mass and a trajectory towards adipose tissue gain [28].
The chronic inflammation seen in PLWHIV not only leads to metabolic alterations but also directly affects bone health. ART, particularly protease inhibitors, exacerbates these issues by inducing lipodystrophy and increasing the production of inflammatory cytokines such as IL-6 and TNF-α. This inflammation interferes with normal insulin receptor function, worsening insulin resistance and contributing to reduced bone mineral density (BMD) [89].
CVD, insulin resistance, diabetes, hypertension, and NAFLD have all been individually linked to low bone turnover and osteoporosis [92,93,94,95,96]. Perimenopausal women have been shown to experience an increase in fat mass and waist circumference, as well as a decrease in skeletal muscle mass [97]. A study that analysed perimenopausal women over a period of six years demonstrated a cumulative increase of 3.4 kg of fat, a 5.7 cm increase in waist circumference, and a 0.23 kg reduction in skeletal mass [97]. These changes were associated with an increase in follicle-stimulating hormone (FSH) during the peri-menopause transition [97]. These modifications experienced by women during the menopause period have been linked to dyslipidaemia, an increased risk of CVD, and insulin resistance [98]. Similar changes are seen in PLWHIV with lipodystrophy, which affects up to 50% of patients. This syndrome is associated with a redistribution of fat, particularly an increase in visceral fat and a reduction in subcutaneous fat, alongside insulin resistance, which not only heightens cardiovascular risk but also contributes to bone deterioration through mechanisms such as increased osteoclast activation [83,89].
A similar experience of fat re-distribution is experienced by PLWHIV, known as lipodystrophy syndrome, where there is an increase in truncal adiposity and a decrease in muscle mass [99]. Up to 50% of all PLWHIV experience such changes [99]. WLHIV have been shown to be more likely to undergo an increase in truncal visceral adipose tissue as compared to non-infected women (age range: 33–45 years) [100]. Moreover, high levels of total visceral adipose tissue have been linked to higher triglyceride levels and lower high-density lipoprotein (HDL) levels in WLHIV [101].
Importantly, physical activity has been shown to improve bone mass in PLHIV and is encouraged in clinical settings. A recent randomised control trial (RCT) demonstrated that combined resistance and aerobic training led to significant increases in BMD in the lumbar spine, femoral neck, and radius (all p < 0.05). Therefore, post-menopausal WLHIV should be encouraged to engage in exercise to mitigate bone loss [102].
Notably, the gut microbiome imbalance in people living with HIV (PLWHIV) has been shown to contribute to insulin resistance, dyslipidaemia, and chronic low-grade inflammation, which are central components of metabolic syndrome. This microbial dysbiosis alters the production of short-chain fatty acids (SCFAs) and bile acid metabolism, leading to increased lipogenesis (fat formation) and impaired glucose regulation. The combination of microbial translocation from the gut and systemic inflammation also exacerbates insulin resistance and promotes the development of atherosclerosis, increasing the risk of cardiovascular diseases in PLWHIV [103].
Non-alcoholic fatty liver disease (NAFLD). A cross-sectional study of 89 PLWHIV on TDF-based cART for over three years found that patients with NAFLD had worse bone mineral density (BMD) than those without NAFLD. The rates of osteopenia (42.86% vs. 25.93%) and osteoporosis (17.14% vs. 3.70%) were significantly higher in PLWHIV with NAFLD. After adjusting for confounders, the odds ratio for poorer BMD in NAFLD patients was 4.49 (95% CI 1.42, 14.15), suggesting a strong association between NAFLD and decreased BMD in PLWHIV [104].
Only a limited number of studies have exclusively analysed the risk of metabolic diseases in WLHIV. After accounting for age, ethnicity, and weight, WLHIV were found to have higher levels of triglyceride, C-reactive protein (CRP), insulin, IL-6, and adiponectin compared to non-HIV infected women [105]. Following the collection of both male and female data, one study exclusively analysed the female population for the risk of presentation with a myocardial infarction (MI). It revealed that female subjects exhibited acute MI rates that were significantly higher (p < 0.0001) in WLHIV compared to non-HIV-infected women [86]. Furthermore, the study also demonstrated that the rate of MI increased considerably with age [86], thus suggesting that menopausal WLHIV are at a higher risk of presenting with an MI.
As PLHIV are already at a higher risk of developing metabolic diseases, post-menopausal WLHIV are at an additionally higher risk of developing metabolic disease due to the superimposed increased risk of developing co-morbidities because of age and hormonal changes, which may subsequently lead to bone loss [78].
Nutritional deficiencies (vitamin D and calcium). Vitamin D is crucial in calcium absorption in the gut, which maintains normal bone homeostasis [106]. Deficiency in vitamin D is defined as serum 25-OH vitamin D < 50 nmol/L (20 ng/mL) [107]. Between 24 and 81% of PLHIV are deficient in vitamin D [108]. The variance in reporting is due to differences in cut-offs used to define deficiency in vitamin D, different geographical locations, varying proportions of patients with darker skin tones, and differences in cART use [109]. A recent meta-analysis of 15 studies revealed that PLWHIV are more likely to be deficient in vitamin D as compared to the general population [110]. Increasing age, male sex, lower BMI, and treatment with cART were shown to be risk factors for vitamin D deficiency [110]. Patients with darker skin tones and of African descent have been shown to have lower vitamin D levels [111]. The literature suggests that despite abundant sunlight, vitamin D deficiency is prevalent in tropical regions due to several factors. Urban residents often spend more time indoors, reducing sun exposure, and air pollution blocks UVB rays needed for vitamin D synthesis. Cultural practices, such as wearing body-covering clothing, limit skin exposure, particularly in Muslim-majority areas. Additionally, people with darker skin tones have more melanin, which lowers vitamin D production [112,113,114].
Moreover, traditional risk factors, such as malabsorption due to HIV-related changes in the gastrointestinal tract, obesity, decreased mobility, and sun exposure, have been linked to lower vitamin D [110]. cART regimes, including efavirenz and protease inhibitors, as well as chronic inflammation due to HIV infection, have been linked to a decrease in vitamin D [115]. Hip and spine BMD reductions were less in patients on cART who were treated with calcium (1000 mg/day) and high-dose vitamin D (4000 IU) at the start of an efavirenz-/emtricitabine-/TDF-based regimen compared to the placebo group [109]. This finding highlights the necessity of stopping bone demineralisation and treating osteoporosis in PLWHIV. Notably, despite efavirenz’s usage, the positive impact of vitamin D supplementation was seen [109].
Recent studies have shown that vitamin D supplementation, particularly in high doses, can have a positive impact on BMD in HIV patients. High-dose vitamin D supplementation (e.g., 20,000 IU weekly) combined with calcium has been shown to reduce musculoskeletal complications and improve overall bone health in patients on antiretroviral therapy (ART), particularly those taking tenofovir or efavirenz, which are known to negatively affect bone metabolism [116]. Another study confirms that vitamin D supplementation can improve BMD, especially in those with significant vitamin D deficiency, although outcomes depend on the dosing regimen and duration of supplementation [117]. In contrast, low-dose supplementation (e.g., 800 IU/day) has been less effective in significantly improving BMD, even though it raises serum vitamin D levels [118]. Furthermore, longer intervention periods (beyond six months) are generally required to see meaningful changes in BMD [117]. In some cases, combining vitamin D with other bone-protective therapies, such as bisphosphonates, might be necessary to achieve optimal results [116,118].
HIV and undernutrition have a reciprocal link. They have been widely documented in PLWHIV [119]. Both play a role in the increasing deterioration of the immune system [119]. HIV diminishes adequate nutritional status, and inadequate nutrition weakens PLWHIV immune systems [119]. Poor nutrition leads to a decrease in minerals such as magnesium, phosphorus, and calcium, which are the primary building blocks of teeth. According to Wadhwa et al., WLHIV are likely to have fewer teeth (p = 0.001), thus feeding into the cycle of poor nutritional intake [120].
Thyroid disease in HIV patients. HIV and thyroid disease leading to bone loss HIV infection can impact the endocrine system, including thyroid function, through direct and indirect mechanisms [121]. The virus itself, cART, and the chronic inflammatory state induced by HIV all contribute to potential thyroid dysfunction [85]. Common thyroid abnormalities in HIV-infected individuals include subclinical hypothyroidism, overt hypothyroidism, euthyroid sick syndrome, overt hyperthyroidism, and Grave’s disease [122]. cART, while lifesaving, has been implicated in both the development and exacerbation of thyroid disorders due to its effects on immune function [121].
In HIV patients, both subclinical and overt hypothyroidism are common, with an increased prevalence linked to chronic inflammation and antiretroviral therapy. Hypothyroidism slows down bone remodelling, causing older bone tissue to accumulate, which can lead to reduced bone density and an increased risk of fractures. In subclinical hypothyroidism, particularly in women, reduced BMD in the hip and spine has been reported, increasing the risk of osteoporosis [123,124].
HIV patients, particularly those with hyperthyroidism, are at risk of accelerated bone resorption, where bone breakdown occurs faster than bone formation. This process, exacerbated by the increased metabolic demands of hyperthyroidism, leads to significant bone loss and a heightened risk of osteoporosis and fractures. Subclinical hyperthyroidism, with suppressed TSH levels, also predisposes patients to fractures, especially in the femoral neck [123,124].
Grave’s disease: This autoimmune condition, seen in PLWHIV due to immune system dysregulation, accelerates bone turnover, leading to osteopenia and osteoporosis. Excessive thyroid hormone levels increase osteoclast activity (bone resorption), contributing to substantial bone loss and an increased risk of fragility fractures [124].
Thyroxine-binding globulin (TBG) levels have been shown to be elevated in PLWHIV and show inverse correlation with CD4+ counts [125]. Patients with a longer HIV diagnosis have most frequently exhibited hypothyroidism [126]. Low levels of tetraiodothyronine (T4), triiodothyronine (T3), and reverse triiodothyronine (rT3) have been shown in advanced HIV disease [127]. However, the elevated reverse T3 levels characteristic of euthyroid sick syndrome have not been linked to advanced HIV disease [127]. This relationship becomes even more complex in post-menopausal women, who already experience higher incidences of hyper- and hypothyroid disease due to a decline in oestrogen levels, which affects the levels of TBG and, in turn, free thyroid hormone levels [128].
Both hyper- and hypothyroidism lead to a net loss of bone. Overt hypothyroidism causes decreased osteoclastic and osteoblastic activity, leading to a prolonged bone remodelling cycle [129]. In contrast, hyperthyroidism leads to an accelerated bone remodelling cycle, which prevents the adequate formation of new bone cells [129]. T3 has been directly linked to the action of osteoblasts on chondrocytes. However, its effect on osteoclasts remains unclear [130]. Thyroid disease in PLWHIV has been linked to the chronic stress response as a result of prolonged viral infection that intricately impacts the hosts’ immune systems [127]. However, the associated co-morbidities of HIV infection and its treatments have shown clearer mechanisms of thyroid dysfunction. Opportunistic pathogens and neoplasms can invade endocrine organs and cause hormone dysfunction. For example, Kaposi’s sarcoma (KS) and lymphoma can infiltrate the thyroid gland, leading to destruction (and subsequent hypothyroidism) or enlargement [131]. In particular, Pneumocystis jiroveci and Cryptococcus neoformans can invade the thyroid gland and cause inflammatory thyroiditis, resulting in either hyper- or hypothyroidism [132]. Moreover, some cART regimes can complicate thyroid dysfunction further.
Conditions such as subclinical hypothyroidism and overt hyperthyroidism have been reported more frequently in patients on long-term ART. Specifically, older regimens, such as those including efavirenz and nevirapine, are more likely to be associated with these thyroid abnormalities. The chronic inflammation caused by HIV, combined with ART, disrupts the endocrine system, leading to thyroid issues, which in turn can exacerbate bone loss due to impaired calcium metabolism [133]. While access to newer ART drugs, such as dolutegravir, is increasing, the use of older and more accessible regimens in low-income countries continues to be a factor in these thyroid-related complications. Studies suggest that thyroid dysfunction contributes to the metabolic and bone health issues commonly observed in PLWHIV in these regions, making it essential to monitor thyroid function as part of comprehensive HIV care [134].
For example, rifampin, phenytoin, ketoconazole, and ritonavir can induce hepatic microsomal enzymes and cause alterations in the clearance of thyroid hormones [135]. A recognised treatment of KS, interferon-Alpha, has also been shown to cause autoimmune thyroiditis [135].
The intricate links between menopause and thyroid dysfunction leading to bone loss in WLHIV have not been explored in clinical studies. However, a meta-analysis study exploring the outcomes of patients with differentiated thyroid carcinoma demonstrated that post-menopausal women in the general population experience significantly greater loss of BMD compared to pre-menopausal women and men [136]. This may demonstrate that the additional immunological burden of HIV in post-menopausal women and the increased risk of thyroid dysfunction due to multi-fold pathology may lead to increased loss of BMD.
Hepatitis C virus (HCV) in HIV patients. Approximately 6.2% of all PLWHIV worldwide are co-infected with HCV [137]. HCV coinfection, for example, is linked to a 1.6-fold higher risk of fractures due to its detrimental impact on bone health and accelerated liver disease progression [138,139]. This has been corroborated by two meta-analyses that have consistently shown that HCV co-infection increases the risk of osteoporosis in PLWHIV. Dong et al. evidenced that the pooled incidence risk ratio of overall fracture risk in HIV/HCV con-infected individuals was 1.77 (95% CI 1.44–2.18) [139]. Similarly, O’Neill et al. showed that low BMD was increasingly prevalent among co-infected patients compared to HIV mono-infected controls (pooled OR 1.98, 95% CI 1.18, 3.31) [140]. A study in Taiwan analysed a total of 636 menopausal women aged 45–80 years using multivariable regression analysis to explore the link between menopause, BMD, and HCV infection [141]. It revealed that BMD was significantly lower in the HCV-seropositive participants in different anatomic locations than in seronegative individuals (lumbar spine: −1.5 vs. −1.1; total hip: −0.9 vs. −0.6; femoral neck: −1.2 vs. −1.0; p < 0.05). HCV-seropositive subjects had higher rates of major osteoporotic fractures (11.3% ± 7.6%vs 9.0 ± 6.8%; p < 0.001) [141]. These findings may be explained by González-Reimers et al.’s study, who found that sclerostin levels are higher in chronic HCV-infected post-menopausal patients with lower BMD [142]. Secreted by osteocytes, sclerostin is negatively correlated with bone turnover and osteoporosis in post-menopausal women [143]. Carlo et al. recently concluded disease progression in HIV/HCV co-infection is impacted by vitamin D deficiency and is a risk factor for osteoporosis and resultant bone fractures [144]. Moreover, the interaction between the immune system and the two viruses may cause the upregulation of RANKL and OPG pathways, resulting in bone loss [104,105].
Bone loss due to use of cART. PLWHIV who are being treated with cART have an increased prevalence of reduced bone mineral density and osteoporosis than untreated HIV patients [106,107,108]. Along the same vein, individuals with HIV and HCV co-infection have been shown to have low Vitamin D levels, thus leading to an increased risk of bone loss [145]. Increased RANKL-mediated bone resorption may lead to a sudden decrease in BMD and lead to the resultant increased prevalence of osteoporosis in PLHIV [146,147,148]. Long-term TDF therapy in HIV and HBV infections is associated with increased loss of BMD [149]. Switching to a non-TDF-based regimen can improve BMD [149]. Decreases in BMD have also been corroborated in HIV-negative patients who take TDF for HIV prevention prophylaxis [111]. The rate of bone loss is the highest during the first one to two years after cART initiation and approaches the degree observed in patients using other medications, such as aromatase inhibitors or glucocorticoids [148].
Efavirenz can lead to decreased serum vitamin D levels via its effects on vitamin D metabolism and binding proteins, leading to bone loss [148]. However, with long-term cART and viral suppression, BMD may increase and stabilise over time [148]. TDF has been shown to cause higher loss in BMD as compared to other types of ART [150]. Cidofovir, an antiviral used to treat cytomegalovirus (CMV) infection with viremia, can cause proximal renal tubulopathy with resulting hypocalcaemia, hypophosphatemia and greater decrease in [151]. Foscarnet, another antiviral also used to treat CMV, can cause hypocalcaemia, hypomagnesaemia, and hypokalaemia through its effects on renal tubular absorption [152]. The combination of foscarnet and pentamidine, a medication used to prevent and treat P. jiroveci pneumonia, can cause severe hypocalcaemia [153].
AIDS-related lymphoma, certain Mycobacterial strains, or C. neoformans infections can cause hypercalcemia due to increased extrarenal activation of vitamin D by activated macrophages or tumour cells [154]. Occasionally, hypercalcemia may occur after initiating cART as part of an immune reconstitution syndrome [154].
A systematic review and meta-analysis highlighted significant bone mineral density (BMD) loss in people living with HIV (PLHIV) following the initiation of antiretroviral therapy (ART). Within the first year of ART, average BMD reductions were 2.68% at the lumbar spine (LS) and 2.66% at the total hip (TH). Notably, the analysis showed that tenofovir disoproxil fumarate (TDF) was associated with more substantial BMD loss compared to tenofovir alafenamide (TAF). Over 48 and 96 weeks, TAF caused less BMD decline, with reductions of 1.57% to 1.90% at the LS and 2.00% to 2.66% at the TH, whereas TDF caused significantly more BMD loss. These findings suggest that switching from TDF to TAF may improve bone health outcomes in PLHIV. The study also found that protease inhibitors (PIs) and ART-naïve patients initiating TDF treatment experienced greater BMD losses, indicating that early ART decisions significantly impact long-term bone health [155]. While the adverse effects of specific drugs such as TDF on bone health are well established, there is significant variability in the degree of bone loss associated with different drug classes within cART regimens. To provide a clearer understanding of the relationship between these drug classes and their specific mechanisms of bone loss, Table 2 outlines key findings from recent studies (Figure 2).

3.2.2. Clinical and Diagnostic Assessment

Post-menopausal WLHIV face unique clinical challenges that require specialised medical attention. When they present to hospitals, these women exhibit a combination of complications arising from both their HIV status and menopausal changes [116]. They are more likely to present with co-morbidities that lead to loss of bone minerals, such as metabolic dysfunction, including diabetes mellitus, insulin resistance, and CVD [158]. Such individuals are also more likely to present with HCV co-infection. Additionally, vitamin D deficiency, prevalent among PLWHIV, significantly contributes to bone health problems, as discussed above [159,160].
Social factors also play a crucial role in bone health management. Women living in different strata of socioeconomic accessibility may experience a variety of barriers when accessing treatment, including transport to the clinic/hospital, the cost of treatment, the inability to attend appointments due to inflexible shift patterns at work, a loss of income due to disability, social stigma, and a lack of awareness of the importance of clinical treatment and follow-up.
PLWHIV have historically struggled with modifiable risk factors such as smoking, alcohol, and drug addiction, thereby increasing the risk of bone loss [161]. Smoking and chronic alcohol use are especially detrimental, reducing BMD and increasing the risk of fractures by approximately 60% compared to non-smokers [162,163]. Addressing these issues requires clinicians to take a holistic approach, including an understanding of addiction, awareness of local support groups who specialise in work with PLWHIV, and a culturally sensitive manner of addressing these unique issues. Table 3 summarises the key modifiable risk factors that clinicians should pay attention to and aim to address during patient contact.
Effective screening and diagnostic tools are essential for monitoring and managing bone density in WLHIV. DXA scans are the gold standard for measuring BMD [168]. The FRAX tool evaluates fracture risk but is likely to underestimate it in HIV-positive patients, as it does not account for HIV-specific factors, so utility of DEXA scans may be more accurate in these instances [109].
Studies have found that the FRAX score often underestimates fracture risk in this population [169,170,171,172]. This is largely due to the tool not accounting for HIV as a secondary cause of osteoporosis. PLWH often have an increased prevalence of bone mineral density (BMD) loss due to long-term antiretroviral therapy (ART) and an over-representation of traditional risk factors, such as smoking, vitamin D deficiency, and low body mass index (BMI) [170,171,172]. For instance, Mazzitelli et al. (2021) found that only a small fraction of PLWH with DXA-confirmed osteoporosis had FRAX scores above the 10% threshold, showing poor sensitivity in identifying high-risk individuals [172]. One of the key shortcomings of FRAX in this context is its lack of consideration for the unique biological and clinical factors affecting PLWH. ART, particularly treatments involving tenofovir and protease inhibitors, accelerates bone loss, and HIV itself may exacerbate bone demineralisation through chronic immune activation and inflammation [172]. McGee and Cotter (2023) noted that even when HIV-related factors are included, the FRAX tool does not adequately reflect the true fracture risk unless BMD data are used. However, in resource-limited settings where routine BMD testing may not be feasible, relying on FRAX alone could lead to missed diagnoses and inadequate preventive measures for those at higher risk [171].
To address these limitations, researchers have suggested modifications to the FRAX tool or the use of alternative or complementary risk assessment methods. Mazzitelli et al. (2021) proposed incorporating HIV as an automatic risk factor for secondary osteoporosis in FRAX calculations, as well as routinely using BMD data in the assessment [172]. In addition to FRAX, McGee and Cotter (2023) highlighted the importance of integrating tools that assess frailty and fall risk, given that PLWH, particularly those ageing with the condition, often face a higher risk of falls [171]. Furthermore, Vizcarra et al. (2024) emphasised that using complementary measures, such as the trabecular bone score (TBS), could provide more comprehensive insights into bone health by evaluating bone microarchitecture, which is not captured by FRAX [170].
Ultimately, early screening and intervention are essential to reduce fracture risk in PLWH. Given the limitations of FRAX, a more holistic approach combining multiple tools may be necessary for more accurate risk stratification. For younger individuals and those on long-term ART, routine BMD assessments alongside modified FRAX scores may better identify those needing intervention. Implementing strategies to optimise bone health early, particularly in PLWH, could mitigate the elevated risk of fractures and improve overall quality of life as they age.
WLHIV have lower BMD at the spine, hip, and radius compared to women without HIV, with the largest difference at the spine [148]. The difference in BMD between pre-/early perimenopausal and late peri/post-menopausal women with HIV is larger than in women without HIV, particularly at the radius and spine [173].
The prevalence of vertebral fractures is particularly elevated, with studies indicating an odds ratio of approximately 1.7, translating to a 70% higher risk compared to HIV-negative women [174,175]. Additionally, femoral neck fractures occur in about 18% of HIV-positive post-menopausal women, while spinal fractures are present in 32% of this population [176]. Beyond vertebral and femoral neck fractures, other fragility fractures such as those of the wrist and hip are also more common in HIV-positive post-menopausal women. These women experience an overall fracture incidence rate that is 1.5 to 1.9 times higher than the general population [177]. A study in Spain examined fracture risk in people living with HIV. Out of over 1.1 million participants, 0.22% had HIV/AIDS. HIV increased the risk of hip fractures by almost five times. The fracture risk was higher in those aged 59 and over [178].

3.3. Challenges and Tribulations Facing Post-Menopausal WLHIV in Low-Income Countries (LICs)

Table 4 contains important findings from studies in LICs; the main studies, such as systematic reviews, clinical trials, and main population studies, have been included.
As demonstrated in Table 4, Black WLHIV in South Africa experience lower BMD at critical sites, such as the distal radius, with 2.8% of virologically suppressed Black women developing osteoporosis. Additionally, menopause is significantly linked to lower BMD, particularly in the femoral neck [18]. In Thailand, age and lower BMI are strongly associated with reduced BMD in the lumbar spine, total hip, and femoral neck [35]. In India, menopause is a major factor contributing to low BMD, with earlier menopause and lower T-scores in the lumbar spine and femoral neck being prevalent among Indian WLHIV [155].
Brazilian WLHIV on long-term antiretroviral therapy (ART) show a significantly earlier onset of menopause and higher FRAX scores, indicating an elevated fracture risk [156]. A longitudinal study in Soweto, South Africa, found significant bone loss in post-menopausal WLHIV, with increased osteoporosis risk after adjustments for HIV status [157].
Across sub-Saharan Africa, ethnic-specific FRAX models reveal a high risk of osteoporosis in older WLHIV. Key risk factors include HIV, malnutrition, and chronic inflammation (“inflammaging”). Limited access to diagnostic tools such as DEXA scans and low awareness of bone health issues further exacerbate this problem in resource-limited settings [158,159,160,164,165,166,167].
Post-menopausal bone loss is a significant health concern in LICs. For example, the prevalence of osteoporosis in Africa ranges from 18.2% to 65.8% [189] across sub-Saharan Africa. Studies from Kenya show high rates of osteoporosis among peri-urban women, worsened by limited access to diagnostics and healthcare [190]. Similarly, research from Morocco, Tunisia and sub-Saharan Africa reflects a high prevalence of osteopenia and osteoporosis, with poor nutrition and healthcare access exacerbating the issue [191].
Moreover, the majority of bone mineral density in Africa was measured using ultrasound due to the limited availability of DEXA scans, which may have underestimated the true prevalence of osteoporosis [189]. A recent call to action for osteoporosis research in sub-Saharan Africa has demonstrated that the prevalence of osteoporosis and fragility fracture incidence is higher than previously researched and that there are large gaps in research that consider Africa as a whole [131,132,133,134,135,136,137,138,139].
Most of the HIV research has concentrated on urban areas, particularly in South Africa [23]. This urban-centric focus leaves a substantial gap in understanding the prevalence of HIV and osteoporosis, the associated risk factors, and its impacts in rural regions within Africa and other LICs. Rural populations often have different lifestyle factors, healthcare access issues, and socio-cultural dynamics compared to their urban counterparts, which can influence the onset and progression of post-menopausal osteoporosis [192]. A recent systematic review demonstrated that, in LICs, individuals living in urban areas were likely to have higher BMD [193]. For example, using a Chinese study population of 490 males and 689 females of ages 50–70, Gu et al. showed that urban men and women had significantly higher BMD than their rural counterparts (p < 0.01). This may be likely due to improved access to healthy food, education, and healthcare in urban settings [194]. In contrast, in higher-income countries (HICs), individuals living in rural areas have been found to be more likely to have higher BMD. For example, a Norwegian study consisting of 10,667 male and female participants with ages ranging between 40 and 75 exhibited higher BMD in rural populations compared to urban ones. This may be due to increased physical activity, more individuals within healthy BMI ranges, and access to less processed foods that may contain greater nutritional benefits [195]. Therefore, whether fracture risk is higher in rural or urban areas may vary according to a country’s economic development. LICs may follow the trend of HICs as they experience continued economic growth.
The lack of comprehensive data from these rural groups hampers the development of tailored interventions that could address their specific needs and improve bone health outcomes for post-menopausal women living in these areas.
WLHIV in rural regions often have low awareness of the importance of treatment and regular health follow-ups for bone health [182]. This lack of knowledge can lead to the delayed diagnosis and treatment of osteoporosis, increasing the risk of fractures and other complications. The healthcare infrastructure in rural areas is typically less developed, with fewer diagnostic facilities available. There are limited specialists available with an interest in bone health. For example, in 2022, there were 30 rheumatologists serving 200 million patients in Nigeria and 2 rheumatologists serving 28 million patients in Ghana [196].
This situation is compounded by logistical challenges such as long distances to healthcare facilities, inadequate transportation options, and difficult road conditions, making it difficult for women to access necessary medical care [197]. Consequently, many women may not receive timely interventions that could prevent or mitigate bone loss. Moreover, different WLHIV may have access to different health resources in neighbourhoods, leading to variances in awareness of the importance of medical follow-up and access to further healthcare [198].
In addition to logistical barriers, WLHIV in LICs face significant social and cultural challenges in managing their health. Social stigma and cultural shame associated with seeking medical help for conditions perceived as sex-related, age-related, or non-urgent can discourage women from pursuing treatment [199]. The cultural stigma surrounding HIV and conditions perceived as sex-related or age-related discourages many women from seeking necessary medical care [200,201].
Compounding these barriers is the prevalent use of traditional medicine, which often coexists with modern healthcare practices. Some communities rely on traditional healers, which can delay or prevent the uptake of modern medical treatments, including antiretroART [202]. Furthermore, responsibilities such as childrearing and household management, often combined with agricultural or informal work, limit women’s time and resources spent on their health [203]. Communities in LICs often depend on locally sourced foods, which can provide nutritional benefits that help manage HIV. However, the availability and variety of these foods are often insufficient to meet the nutritional needs of WLHIV [202].
The economic impact of losing work time to seek medical care can be substantial, further deterring women from accessing healthcare services. A recent cross-sectional study set in South Africa consisting of 996 participants found that individuals in rural areas were more likely to ask relatives and neighbours for assistance with money and food compared to their urban counterparts [204]. These multifaceted barriers highlight the need for targeted awareness campaigns, community-based interventions, and healthcare policies that address the unique challenges faced by post-menopausal women in LICs.
Research suggests that built environments (spaces in which people live and work) influence health behaviours [205]. The resources and road conditions in the neighbourhood can give residents the opportunity to seek medical attention, and this leads to more accessibility of HIV testing facilities and more screening and early detection of HIV and the prevention of complications [206]. It is crucial to remember that stigma and discrimination related to HIV may discourage locals from utilising adjacent HIV health services [207]. Studies have shown that, in LICs, poor road conditions and limited transportation options create significant barriers for people, particularly WLHIV, to reach healthcare facilities. Proximity to healthcare centres is directly associated with higher rates of HIV testing and treatment adherence, while more distant or harder-to-reach clinics are linked to lower service utilisation and delayed diagnosis [208]. Additionally, community characteristics, such as the availability of healthcare resources and social support networks within neighbourhoods, have been found to significantly influence the accessibility of HIV services. Areas with better healthcare infrastructure, including more accessible HIV testing facilities, foster earlier detection of HIV, improved ART adherence, and better overall health outcomes [209]. To improve access, targeted interventions such as mobile health services and improving road infrastructure can significantly enhance the ability of people to seek timely medical attention. Public health policies that focus on improving the built environment and increasing the accessibility of health services can make a significant difference in HIV care and prevention in LICs [208,209].
In HICs, diagnostic tools such as DEXA scans are widely accessible, allowing for routine screening of osteoporosis, particularly among at-risk groups such as WLHIV. Early detection is crucial in preventing severe bone loss and reducing fracture risks [210,211]. Conversely, LICs suffer from limited access to advanced diagnostic technologies. Many rural areas in LICs rely on heel quantitative ultrasound, which, although cost-effective, is less accurate and may underestimate the prevalence and severity of bone loss [212,213]. This diagnostic gap highlights the need for portable and affordable diagnostic tools, such as mobile DEXA units, to enhance early detection and intervention in LICs [211].
Heterosexual women have been found to experience greater disclosure shame compared to heterosexual males and non-heterosexual females [214]. Therefore, disclosure of shame may affect healthcare-seeking behaviour in post-menopausal WLHIV. Additionally, a cross-sectional study comparing 64 LICs and middle-income countries found that HIV stigma was lower in countries with greater gross domestic product per capita [215]. HIV stigma was also lower in men and those who had adequate knowledge.
Treatment Options and Availability: HICs benefit from ART, such as tenofovir alafenamide (TAF), which is bone-sparing compared to older regimens such as tenofovir disoproxil fumarate (TDF). This has significantly reduced osteoporosis-related complications in WLHIV in HICs [210,213]. However, in LICs, the continued reliance on TDF due to its lower cost exacerbates bone loss, particularly in older women who are already vulnerable due to HIV-related factors. Additionally, access to osteoporosis treatments such as bisphosphonates, calcium, and vitamin D is more widespread in HICs, further improving bone health outcomes [211,212].
Healthcare Infrastructure: The healthcare systems in HICs are better equipped to manage osteoporosis and other bone health issues in HIV-positive populations. Public health campaigns, regular screenings, and easy access to specialised care ensure that WLHIV receive timely and effective treatment [216]. In contrast, LICs face significant shortages in terms of healthcare providers, particularly specialists such as rheumatologists, resulting in delayed or suboptimal care [213]. Rural populations are particularly disadvantaged due to long distances to healthcare facilities, limited transportation options, and a lack of necessary medical expertise [211,213].
Potential Interventions: Addressing these disparities requires several interventions. First, expanding the availability of bone-sparing ARTs such as TAF in LICs would help reduce bone loss among WLHIV. Additionally, community-based educational programs to raise awareness about osteoporosis, along with mobile healthcare units or telemedicine solutions, could provide critical care to women in rural and underserved regions. Ensuring the availability of diagnostic tools and treatments, such as calcium and vitamin D supplements, would also improve outcomes [210,211,216].

3.3.1. Management

As discussed above, the unique population of post-menopausal WLHIV requires a tailored approach consisting of non-pharmacological and pharmacological management.
Non-pharmacological management: The following aspects of care play a key role in the non-pharmacological management of bone fracture risk in post-menopausal WLHIV:
  • Education and awareness of premature menopause and associated risks of bone loss. Education can help prevent premature menopause by promoting healthier lifestyle choices, such as reducing smoking and improving nutrition, which are key risk factors for early menopause [217]. Educating women about the importance of vitamin D and calcium intake can delay menopause and improve health outcomes. Additionally, increased awareness of early medical intervention options, such as hormone replacement therapy, enables earlier care, reducing the risk of complications from premature menopause [218].
  • Encouragement of regular exercise, particularly resistance and aerobic training [102].
  • Tailored help to address smoking, alcohol, and drug use.
  • Measurement of calcium and vitamin D levels for those at high risk of fracture due to co-morbidities, previous fracture, or family history.
  • Use of DXA scans for all post-menopausal women with regular follow-up [219].
  • Population-specific FRAX tools, in particular for women living in low-income countries [220].
  • Clinical recognition of nutritional requirements of patients in this population–malnourished versus those with diabetes.
  • Social and mental health impact of disease: specialised culturally-sensitive talking services addressed to WLHIV.
  • Joint HIV, bone health, and menopause clinics and MDTs to develop tailored plans.
Pharmacological management: The following aspects of care play a key role in the pharmacological management of bone fracture risk in post-menopausal WLHIV:
  • Personalised cART regimes that balance bone loss at an individual level [221].
  • The addition of bisphosphonates when stopping TDF-based therapy has been shown to increase BMD [222].
  • HRT use has been shown to increase BMD [223].
  • Romosozumab, teriparatide, and denosumab have shown benefit in post-menopausal women [110,153].
  • Abaloparatide (a synthetic peptide analogue of parathyroid hormone-related protein (PTHrP)) is used to manage and treat osteoporosis. Recently, the National Institute for Health and Care Excellence (NICE) recommended abaloparatide as an option for treating osteoporosis in women with menopause, only if they have a very high risk of fracture [153].

3.3.2. Future Directions

Further research is needed to elucidate the intricate interactions between menopause, HIV, and bone health specifically. Currently, most long-term HIV-related studies have explored the impact of the virus on combined populations of males and females, or exclusively males. Most of the HIV literature focused on the female population has explored the transmission and effects of HIV during pregnancy and the postpartum period [101,102]. Therefore, there are stark gaps in the research focused on WLHIV. Long-term longitudinal studies investigating the long-term effects of cART on bone density in post-menopausal women, the impact of lifestyle modification metabolic changes such as fatty liver, obesity, and diabetes on the bone health of post-menopausal WLHIV, and the efficacy of various interventions in this population are crucial, especially with newer potential therapies becoming available, such as abaloparatide [153,154,155]. Additionally, exploring the genetic and molecular mechanisms underlying the susceptibility to bone loss in HIV-positive post-menopausal women could pave the way for personalised therapeutic approaches, as menopause is a pivotal life event and is experienced in a unique manner by individuals. Furthermore, studies investigating the social and mental health impact of early menopause and fractures in WLHIV need to take place [156]. As we suggested before, metabolic clinics for PLWHIV represent an essential part in the treatment of cardiovascular disease and metabolic bone disease [157,164].
Future research should be more targeted and specific. To address current knowledge gaps, long-term cohort studies are essential. These should focus on the impact of different antiretroviral therapies, such as comparing bone-sparing therapies such as tenofovir alafenamide (TAF) with older regimens, such as tenofovir disoproxil fumarate (TDF), in WLHIV. This would help identify optimal treatments to minimise bone loss.
Additionally, intervention trials should examine the effects of lifestyle changes, such as diet, physical activity, and management of metabolic conditions such as diabetes or obesity, on bone density in this population. Such studies should evaluate both pharmacological and non-pharmacological strategies, potentially testing newer drugs such as abaloparatide alongside lifestyle modifications.
Genetic and molecular research is another priority area, with studies needed to explore how HIV accelerates bone loss during menopause. These findings could guide the development of personalised therapies. Lastly, qualitative research into the mental and social impacts of early menopause and fractures in WLHIV is important to inform holistic care approaches, including the integration of bone health services in HIV care clinics.
Research should further explore the role of nutrition and access to nutrient-dense foods in this population, particularly given that HIV-related gastrointestinal complications and nutrient malabsorption can worsen deficiencies in essential nutrients, such as calcium, vitamin D, and others vital for bone health. Incorporating access to locally sourced, nutritious foods and educating WLHIV on nutritional strategies could be an integral part of their HIV care.

3.3.3. Limitations and Strengths

The limitations of our narrative review include the broad search across different research databases. This is not a systematic review or meta-analysis, as the focus is on the entire topic, which may lead to some variation in the types of studies included. Another limitation is that we have only included studies published in the English language. The strengths of our review include the use of a variety of studies from different geographical locations, allowing for a richer and more nuanced perspective on the topic. This review sets the stage for current research evidence available on post-menopausal women living with HIV (WLHIV) who are at risk of bone fractures. It has allowed specific gaps in the literature to be identified and has provided direction for future research. Importantly, the review covers data from both developed and developing countries, enabling health professionals in developing countries to establish health policies that will help narrow the gap in terms of health services. A unique strength of this review is that it provides robust evidence for why “an initiative to promote the concept of effective menopausal care among women living with HIV” is needed in order to improve bone health. Additionally, the narrative style of the review may make it useful for a wider audience, including clinicians, policymakers, postgraduate and undergraduate students, and laypeople.

4. Conclusions

Menopause and HIV infection independently contribute to significant bone loss and increased fracture risk. The convergence of these conditions in post-menopausal women living with HIV exacerbates bone health issues, necessitating a multifaceted approach to management. Optimising cART regimens, implementing lifestyle modifications, and providing individuals with tailored clinical care are key strategies to mitigate the adverse effects on bone health. Ongoing research and tailored interventions will be essential in improving outcomes for this vulnerable population.

Author Contributions

Conceptualization, M.H.A. and A.H. (Ahmed Hassan), methodology, M.J. and A.H. (Amirmohammad Heidari), software, M.J. and A.H. (Amirmohammad Heidari); validation, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; formal analysis, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; investigation, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; resources, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; data curation, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; writing—original draft preparation, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; writing—review and editing, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; visualization, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; supervision, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; project administration, M.J., A.H. (Amirmohammad Heidari), A.H. (Ahmed Hassan), D.M., O.P., M.P. and M.H.A.; funding acquisition, M.H.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

This review article and all information available included in this review.

Acknowledgments

Authors would like to thank their families for their support during preparation of this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lima, V.D.; Hogg, R.S.; Harrigan, P.R.; Moore, D.; Yip, B.; Wood, E.; Montaner, J.S.G. Continued improvement in survival among HIV-infected individuals with newer forms of highly active antiretroviral therapy. AIDS 2007, 21, 685–692. [Google Scholar] [CrossRef] [PubMed]
  2. Losina, E.; Freedberg, K.A. Life expectancy in HIV. BMJ 2011, 343, 7829. [Google Scholar] [CrossRef] [PubMed]
  3. Sabin, C.A. Do people with HIV infection have a normal life expectancy in the era of combination antiretroviral therapy? BMC Med. 2013, 11, 251. [Google Scholar] [CrossRef] [PubMed]
  4. Loutfy, M.; Andany, N.; Kennedy, V.L.; Aden, M. Perspectives on menopause and women with HIV. Int. J. Womens Health 2016, 8, 1–22. [Google Scholar] [CrossRef]
  5. UNAIDS. Global HIV and AIDS Statistics—Fact Sheet; UNAIDS: Geneva, Switzerland, 2024; Available online: https://www.unaids.org/sites/default/files/media_asset/UNAIDS_FactSheet_en.pdf (accessed on 17 August 2024).
  6. AARP International. How Women 50-Plus Are Driving the Global Longevity Economy; AARP: Washington, DC, USA, 2020. [Google Scholar]
  7. Gao, T.Y.; Zhao, L.K.; Liu, X.; Li, H.Y.; Ma, Y.T.; Fang, W.; Wang, X.; Zhang, C. Disease burden of AIDS in last 30-year period and its predicted level in next 25-years based on the global burden disease 2019. BMC Public Health 2024, 24, 2384. [Google Scholar] [CrossRef]
  8. UNAIDS. HIV and adolescent girls and young women—Thematic Briefing Note—2024 Global AIDS update The Urgency of Now: AIDS at a Crossroads. Available online: https://www.unaids.org/en/resources/documents/2024/2024-unaids-global-aids-update-adolescent-girls-young-women (accessed on 17 August 2024).
  9. Whiteley, J.; DiBonaventura, M.D.; Wagner, J.-S.; Alvir, J.; Shah, S. The Impact of Menopausal Symptoms on Quality of Life, Productivity, and Economic Outcomes. J. Womens Health 2013, 22, 983–990. [Google Scholar] [CrossRef]
  10. World Health Organisation. WHO 2022—Menopause. Available online: https://www.who.int/news-room/fact-sheets/detail/menopause (accessed on 6 September 2024).
  11. Peacock, K.; Carlson, K.; Ketvertis, K.M. Menopause; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  12. Gehrke, B.; Farias, M.L.F.; Wildemberg, L.E.; Ferraiuoli, G.I.; Ribeiro, V.; Bosgnoli, R.; Neto, F.d.P.P.; de Mendonça, L.M.C.; Madeira, M.; Coelho, M.C.A. Evaluation of bone mineral density, microarchitecture, and detection of fractures on young patients living with human immunodeficiency virus: When and how to screen? Endocrine 2023, 83, 214–226. [Google Scholar] [CrossRef]
  13. Suarez-García, I.; Alejos, B.; Pérez-Elías, M.J.; Iribarren, J.A.; Hernando, A.; Ramírez, M.; Tasias, M.; Pascual, M.; Jarrin, I.; Hernando, V.; et al. How do women living with HIV experience menopause? Menopausal symptoms, anxiety and depression according to reproductive age in a multicenter cohort. BMC Womens Health 2021, 21, 223. [Google Scholar] [CrossRef]
  14. King, E.M.; Swann, S.A.; Murray, M.C.M. Markers of ovarian reserve in women living HIV: A systematic review. HIV Med. 2023, 24, 247–259. [Google Scholar] [CrossRef]
  15. Bedenk, J.; Vrtačnik-Bokal, E.; Virant-Klun, I. The role of anti-Müllerian hormone (AMH) in ovarian disease and infertility. J. Assist. Reprod. Genet. 2020, 37, 89–100. [Google Scholar] [CrossRef]
  16. Zicari, S.; Sessa, L.; Cotugno, N.; Ruggiero, A.; Morrocchi, E.; Concato, C.; Rocca, S.; Zangari, P.; Manno, E.C.; Palma, P. Immune Activation, Inflammation, and Non-AIDS Co-Morbidities in HIV-Infected Patients under Long-Term ART. Viruses 2019, 11, 200. [Google Scholar] [CrossRef] [PubMed]
  17. Abelman, R.; Tien, P.C. The Reproductive Transition: Effects on Viral Replication, Immune Activation, and Metabolism in Women with HIV infection. Curr. HIV/AIDS Rep. 2022, 19, 133–139. [Google Scholar] [CrossRef] [PubMed]
  18. Schank, M.; Zhao, J.; Moorman, J.P.; Yao, Z.Q. The Impact of HIV- and ART-Induced Mitochondrial Dysfunction in Cellular Senescence and Aging. Cells 2021, 10, 174. [Google Scholar] [CrossRef]
  19. Cavalcante, M.B.; Sampaio, O.G.M.; Câmara, F.E.A.; Schneider, A.; de Ávila, B.M.; Prosczek, J.; Masternak, M.M.; Campos, A.R. Ovarian aging in humans: Potential strategies for extending reproductive lifespan. Geroscience 2023, 45, 2121–2133. [Google Scholar] [CrossRef]
  20. Ahmed, M.; Bondje, S.; Jiwan, R.; Rawther, F.; Duku, A.; Husain, N.E.; Woodward, C.; Mital, D. Early menopause in acquired immunodeficiency syndrome. J. Res. Med. Sci. 2021, 26, 122. [Google Scholar] [CrossRef]
  21. Chikwati, R.P.; Jaff, N.G.; Mahyoodeen, N.G.; Micklesfield, L.K.; Ramsay, M.; Gómez-Olivé, F.X.; Mohamed, S.F.; Choma, S.S.R.; George, J.A.; Crowther, N.J. The association of menopause with cardiometabolic disease risk factors in women living with and without HIV in sub-Saharan Africa: Results from the AWI-Gen 1 study. Maturitas 2024, 187, 108069. [Google Scholar] [CrossRef]
  22. Shuster, L.T.; Rhodes, D.J.; Gostout, B.S.; Grossardt, B.R.; Rocca, W.A. Premature menopause or early menopause: Long-term health consequences. Maturitas 2010, 65, 161–166. [Google Scholar] [CrossRef] [PubMed]
  23. Breasail, M.Ó.; Gregson, C.L.; Norris, S.A.; Madanhire, T.; Jaff, N.; Crowther, N.J.; Micklesfield, L.K.; Ward, K.A. Menopause is associated with bone loss, particularly at the distal radius, in black South African women: Findings from the Study of Women Entering and in Endocrine Transition (SWEET). Bone 2022, 164, 116543. [Google Scholar] [CrossRef]
  24. Genant, H.K.; Cooper, C.; Poor, G.; Reid, I.; Ehrlich, G.; Kanis, J.; Nordin, B.E.; Barrett-Connor, E.; Black, D.; Bonjour, J.P.; et al. Interim Report and Recommendations of the World Health Organization Task-Force for Osteoporosis. Osteoporos. Int. 1999, 10, 259–264. [Google Scholar] [CrossRef]
  25. Amarnath, S.S.; Kumar, V.; Das, S.L. Classification of Osteoporosis. Indian J. Orthop. 2023, 57 (Suppl. S1), 49–54. [Google Scholar] [CrossRef]
  26. Melton, J.L. Epidemiology of Spinal Osteoporosis. Spine 1997, 22, 2S–11S. [Google Scholar] [CrossRef] [PubMed]
  27. Downey, C.; Kelly, M.; Quinlan, J.F. Changing trends in the mortality rate at 1-year post hip fracture—A systematic review. World J. Orthop. 2019, 10, 166–175. [Google Scholar] [CrossRef] [PubMed]
  28. Dragovic, B.; Rymer, J.; Nwokolo, N. Menopause care in women living with HIV in the UK—A review. J. Virus Erad. 2022, 8, 100064. [Google Scholar] [CrossRef]
  29. Zahn, K.; Pittman, A.; Conklin, J.; Knittel, A.; Neal-Perry, G. Disparities in menopausal care in the United States: A systematic review. Maturitas 2024, 186, 108021. [Google Scholar] [CrossRef] [PubMed]
  30. Scofield, D.; Moseholm, E.; Aebi-Popp, K.; Hachfeld, A. Management of menopause in women living with HIV—A comparative guideline review. Maturitas 2024, 183, 107937. [Google Scholar] [CrossRef]
  31. Kharsany, A.B.M.; Karim, Q.A. HIV Infection and AIDS in Sub-Saharan Africa: Current Status, Challenges and Opportunities. Open AIDS J. 2016, 10, 34–48. [Google Scholar] [CrossRef]
  32. Joint United Nations Programme on HIV/AIDS. Dangerous Inequalities: World AIDS Day Report; UNAIDS: Geneva, Switzerland, 2022; Available online: https://www.unaids.org/en/resources/documents/2022/dangerous-inequalities (accessed on 17 August 2024).
  33. AA World. Global Population Tops 8 Billion as Aging Trend Accelerates. Turkey. 2024. Available online: https://www.aa.com.tr/en/world/global-population-tops-8-billion-as-aging-trendaccelerates/3271467#:~:text=The%20global%20population%20aged%2065,already%20surpassed%208%20billion%20people (accessed on 17 August 2024).
  34. United States. Gilead Sciences. Gilead’s HIV Age Positively. Phase Two 18-Month Progress Report. 2023. Available online: https://www.gilead.com/-/media/files/pdfs/other/gilead-agepositively-boardreport.pdf (accessed on 17 August 2024).
  35. Tariq, S.; Burns, F.M.; Gilson, R.; Sabin, C. PRIME (Positive Transitions Through the Menopause) Study: A protocol for a mixed-methods study investigating the impact of the menopause on the health and well-being of women living with HIV in England. BMJ Open 2019, 9, e025497. [Google Scholar] [CrossRef]
  36. Manolagas, S.C. Birth and Death of Bone Cells: Basic Regulatory Mechanisms and Implications for the Pathogenesis and Treatment of Osteoporosis. Endocr. Rev. 2000, 21, 115–137. [Google Scholar]
  37. Eisman, J.A.; Bogoch, E.R.; Dell, R.; Harrington, J.T.; McKinney, R.E.; McLellan, A.; Mitchell, P.J.; Silverman, S.; Singleton, R.; Siris, E.; et al. Making the first fracture the last fracture: ASBMR task force report on secondary fracture prevention. J. Bone Miner. Res. 2012, 27, 2039–2046. [Google Scholar] [CrossRef]
  38. Cortés, Y.I.; Yin, M.T.; Reame, N.K. Bone Density and Fractures in HIV-infected Postmenopausal Women: A Systematic Review. J. Assoc. Nurses AIDS Care 2015, 26, 387–398. [Google Scholar] [CrossRef]
  39. Sharma, A.; Hoover, D.R.; Shi, Q.; Tien, P.C.; Weber, K.M.; Shah, J.G.; Yin, M.T. Human Immunodeficiency Virus (HIV) and Menopause Are Independently Associated with Lower Bone Mineral Density: Results from the Women’s Interagency HIV Study. Clin. Infect. Dis. 2022, 75, 65–72. [Google Scholar] [CrossRef] [PubMed]
  40. Leite-Silva, P.R.; Pinheiro, R.S.; Barbosa-Ferreira, J.M.B.; Balieiro, A.; Sabidó, M.; Lacerda, M.V.G.; Chaves, Y.O.; Nogueira, P.A.; Benzaken, A.S. Bone mass, fracture risk, and associated factors in postmenopausal women living with HIV. Menopause 2024, 31, 46–51. [Google Scholar] [CrossRef] [PubMed]
  41. Matovu, F.K.; Pettifor, J.M.; Compston, J.E. HIV and Bone Health Considerations for Menopausal Women Living with HIV in sub-Saharan Africa. J. Bone Miner. Res. 2020, 38, 617–618. [Google Scholar] [CrossRef]
  42. Cabrera, D.M.; Cornejo, M.P.; Slotkin, R.; Pinedo, Y.; Yu, W.; Guan, W.; Garcia, P.J.; Hsieh, E. Prevalence of and risk factors for vertebral fracture and low bone mineral density among Peruvian women aging with HIV. Arch. Osteoporos. 2023, 18, 64. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, T.; Wilson, I.B.; Zullo, A.R.; Meyers, D.J.; Lee, Y.; Daiello, L.A.; Kim, D.H.; Kiel, D.P.; Shireman, T.I.; Berry, S.D. Hip Fracture Rates in Nursing Home Residents with and without HIV. J. Am. Med. Dir. Assoc. 2022, 23, 517–518. [Google Scholar] [CrossRef] [PubMed]
  44. Ilha, T.A.S.H.; Comim, F.V.; Copes, R.M.; Compston, J.E.; Premaor, M.O. HIV and Vertebral Fractures: A Systematic Review and Metanalysis. Sci. Rep. 2018, 8, 7838. [Google Scholar] [CrossRef]
  45. Güerri-Fernandez, R.; Vestergaard, P.; Carbonell, C.; Knobel, H.; Avilés, F.F.; Castro, A.S.; Noguès, X.; Prieto-Alhambra, D.; Diez-Perez, A. HIV infection is strongly associated with hip fracture risk, independently of age, gender, and comorbidities: A population-based cohort study. J. Bone Miner. Res. 2013, 28, 1259–1263. [Google Scholar] [CrossRef]
  46. ERAnand Scott, L.A.; Harrison, W.J. Hip and Knee Replacement in the HIV positive patient. Malawi Med. J. 2012, 24, 14–16. [Google Scholar]
  47. Akkaya, M.; Buday, Z.; Akcaalan, S.; Linke, P.; Gehrke, T.; Citak, M. In-hospital complications following total knee and hip arthroplasty in patients with human immunodeficiency virus. Jt. Dis. Relat. Surg. 2022, 33, 3–8. [Google Scholar] [CrossRef]
  48. Curlewis, K.; Leung, B.; Sinclair, L.; Thornhill, C.; Chan, G.; Ricketts, D. Systemic medical complications following joint replacement: A review of the evidence. Ann. R. Coll. Surg. Engl. 2023, 105, 191–195. [Google Scholar] [CrossRef]
  49. Weitzmann, M.N.; Ofotokun, I.; Titanji, K.; Sharma, A.; Yin, M.T. Bone Loss Among Women Living With HIV. Curr. HIV/AIDS Rep. 2016, 13, 367–373. [Google Scholar] [CrossRef] [PubMed]
  50. Chen, X.; Wang, Z.; Duan, N.; Zhu, G.; Schwarz, E.M.; Xie, C. Osteoblast–osteoclast interactions. Connect. Tissue Res. 2018, 59, 99–107. [Google Scholar] [CrossRef] [PubMed]
  51. Riggs, B.L.; Khosla, S.; Melton, L.J. Sex Steroids and the Construction and Conservation of the Adult Skeleton. Endocr. Rev. 2002, 23, 279–302. [Google Scholar] [CrossRef]
  52. Khosla, S. Minireview: The OPG/RANKL/RANK System. Endocrinology 2001, 142, 5050–5055. [Google Scholar] [CrossRef]
  53. Indridason, O.S.; Franzson, L.; Sigurdsson, G. Serum osteoprotegerin and its relationship with bone mineral density and markers of bone turnover. Osteoporos. Int. 2005, 16, 417–423. [Google Scholar] [CrossRef]
  54. Pollock, E.; Klotsas, A.E.; Compston, J.; Gkrania-Klotsas, E. Bone health in HIV infection. Br. Med. Bull. 2009, 92, 123–133. [Google Scholar] [CrossRef]
  55. Compston, J. HIV infection and bone disease. J. Intern. Med. 2016, 280, 350–358. [Google Scholar] [CrossRef] [PubMed]
  56. Samji, H.; Cescon, A.; Hogg, R.S.; Modur, S.P.; Althoff, K.N.; Buchacz, K.; Burchell, A.N.; Cohen, M.; Gebo, K.A.; Gill, M.J.; et al. Closing the Gap: Increases in Life Expectancy among Treated HIV-Positive Individuals in the United States and Canada. PLoS ONE 2013, 8, e81355. [Google Scholar] [CrossRef]
  57. Xu, J.F.; Wang, P.C.; Cheng, F. Health related behaviors among HIV-infected people who are successfully linked to care: An institutional-based cross-sectional study. Infect. Dis. Poverty 2020, 9, 28. [Google Scholar] [CrossRef]
  58. Titanji, K.; Vunnava, A.; Sheth, A.N.; Delille, C.; Lennox, J.L.; Sanford, S.E.; Foster, A.; Knezevic, A.; Easley, K.A.; Weitzmann, M.N.; et al. Dysregulated B Cell Expression of RANKL and OPG Correlates with Loss of Bone Mineral Density in HIV Infection. PLoS Pathog. 2014, 10, e1004497. [Google Scholar] [CrossRef]
  59. Vikulina, T.; Fan, X.; Yamaguchi, M.; Roser-Page, S.; Zayzafoon, M.; Guidot, D.M.; Ofotokun, I.; Weitzmann, M.N. Alterations in the immuno-skeletal interface drive bone destruction in HIV-1 transgenic rats. Proc. Natl. Acad. Sci. USA 2010, 107, 13848–13853. [Google Scholar] [CrossRef] [PubMed]
  60. Torres, H.M.; Arnold, K.M.; Oviedo, M.; Westendorf, J.J.; Weaver, S.R. Inflammatory Processes Affecting Bone Health and Repair. Curr. Osteoporos. Rep. 2023, 21, 842–853. [Google Scholar] [CrossRef] [PubMed]
  61. Kelesidis, T.; Moser, C.B.; Johnston, E.; Stein, J.H.; Dube, M.P.; Yang, O.O.; McComsey, G.A.; Currier, J.S.; Brown, T.T. Brief Report: Changes in Plasma RANKL-Osteoprotegerin in a Prospective, Randomized Clinical Trial of Initial Antiviral Therapy: A5260s. J. Acquir. Immune Defic. Syndr. 2018, 78, 362–366. [Google Scholar] [CrossRef]
  62. Ono, T.; Hayashi, M.; Sasaki, F.; Nakashima, T. RANKL biology: Bone metabolism, the immune system, and beyond. Inflamm. Regen. 2020, 40, 2. [Google Scholar] [CrossRef]
  63. Delpino, M.V.; Quarleri, J. Influence of HIV Infection and Antiretroviral Therapy on Bone Homeostasis. Front. Endocrinol 2020, 11, 502. [Google Scholar] [CrossRef]
  64. Lin, W.; Li X fu Ren D cheng Song, M.; Duan, L.; Liu, J.-Z.; Zhan, Z. Administration of zoledronic acid alleviates osteoporosis in HIV patients by suppressing osteoclastogenesis via regulating RANKL expression. Mol. Med. 2021, 27, 19. [Google Scholar] [CrossRef] [PubMed]
  65. Triguero-Martínez, A.; Pardines, M.; Montes, N.; Ortiz, A.M.; Iglesia-Cedeira, A.; Valero-Martínez, C.; Martín, J.; González-Álvarot, I.; Castañeda, S.; Lamana, A. Genetic Variants in RANK and OPG Could Influence Disease Severity and Bone Remodeling in Patients with Early Arthritis. Life 2024, 14, 1109. [Google Scholar] [CrossRef]
  66. Ofotokun, I. Deciphering how HIV-1 weakens and cracks the bone. Proc. Natl. Acad. Sci. USA 2018, 115, 2551–2553. [Google Scholar] [CrossRef]
  67. Titanji, K.; Vunnava, A.; Foster, A.; Sheth, A.N.; Lennox, J.L.; Knezevic, A.; Shenvi, N.; Easley, K.A.; Ofotokun, I.; Weitzmann, M.N. T-cell receptor activator of nucleat factor-kB ligand/osteoprotegein imbalance is associated with HIV-induced bone loss in patients with higher CD4+ T-cell counts. AIDS 2018, 32, 885–894. [Google Scholar] [CrossRef]
  68. Eghbali-Fatourechi, G.; Khosla, S.; Sanyal, A.; Boyle, W.J.; Lacey, D.L.; Riggs, B.L. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J. Clin. Investig. 2003, 111, 1221–1230. [Google Scholar] [CrossRef]
  69. Van Ommen, C.E.; Hsieh, A.Y.Y.; Albert, A.Y.; Kimmel, E.R.; Cote, H.C.F.; Maan, E.J.; Prior, J.C.; Pick, N.; Murray, M.C.M.; CIHR Team on Cellular Aging, HIV Comorbidities in Women, Children (CARMO-Endo) CIHR CTN 277. Lower anti-Müllerian hormone levels are associated with HIV in reproductive age women and shorter leukocyte telomere length among late reproductive age women. AIDS 2023, 37, 769–778. [Google Scholar] [CrossRef] [PubMed]
  70. Nelson, S.M.; Davis, S.R.; Kalantaridou, S.; Lumsden, M.A.; Panay, N.; Anderson, R.A. Anti-Müllerian hormone for the diagnosis and prediction of menopause: A systematic review. Hum. Reprod. Update 2023, 29, 327–346. [Google Scholar] [CrossRef] [PubMed]
  71. Scherzer, R.; Greenblatt, R.M.; Merhi, Z.O.; Kassaye, S.; Lambert-Messerlian, G.; Maki, P.M.; Murphy, K.; Karim, R.; Bacchetti, P. Use of antimüllerian hormone to predict the menopausal transition in HIV-infected women. Am. J. Obstet. Gynecol. 2017, 216, e1–e46. [Google Scholar] [CrossRef]
  72. Hachfeld, A.; Atkinson, A.; Stute, P.; Calmy, A.; Tarr, P.E.; Darling, K.; Flury, B.B.; Polli, C.; Sultan-Beyer, L.; Abela, I.A.; et al. Women with HIV transitioning through menopause: Insights from the Swiss HIV Cohort Study (SHCS). HIV Med. 2022, 23, 417–425. [Google Scholar] [CrossRef] [PubMed]
  73. Jones, T.P.W.; Lampe, F.C.; Arenas-Pinto, A.; Smith, C.; McDonnell, J.; Haddow, L.; Johnson, M.; Yousef, E.; Lascar, M.; Geretti, A.M.; et al. Alcohol, smoking, recreational drug use and association with virological outcomes among people living with HIV: Cross-sectional and longitudinal analyses. HIV Med. 2022, 23, 209–226. [Google Scholar] [CrossRef]
  74. Schoenbaum, E.E.; Hartel, D.; Lo, Y.; Howard, A.A.; Floris-Moore, M.; Arnsten, J.H.; Santoro, N. HIV Infection, Drug Use, and Onset of Natural Menopause. Clin. Infect. Dis. 2005, 41, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
  75. Willems, N.; Lemoine, C.; Liesnard, C.; Gervy, C.; Hien, A.D.; Karama, R.; Somda, I.; Englert, Y. Is ovarian function impaired in HIV patients? A clinical pilot study in Burkina Faso. Rev. Med. Brux. 2013, 34, 397–404. [Google Scholar]
  76. Lipid and Lipoprotein Profile in Menopausal Transition. Effects of Hormones, Age and Fat Distribution. Horm. Metab. Res. 2004, 36, 215–220. [Google Scholar] [CrossRef]
  77. Mesch, V.R.; Siseles, N.O.; Maidana, P.N.; Boero, L.E.; Sayegh, F.; Prada, M.; Royer, M.; Schreier, L.; Benencia, H.J.; Berg, G.A. Androgens in relationship to cardiovascular risk factors in the menopausal transition. Climacteric 2008, 11, 509–517. [Google Scholar] [CrossRef]
  78. Jeong, H.G.; Park, H. Metabolic Disorders in Menopause. Metabolites 2022, 12, 954. [Google Scholar] [CrossRef]
  79. Myerson, M.; Malvestutto, C.; Aberg, J.A. Management of lipid disorders in patients living with HIV. J. Clin. Pharmacol. 2015, 55, 957–974. [Google Scholar] [CrossRef] [PubMed]
  80. Grunfeld, C.; Pang, M.; Doerrler, W.; Shigenaga, J.K.; Jensen, P.; Feingold, K.R. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J. Clin. Endocrinol. Metab. 1992, 74, 1045–1052. [Google Scholar] [PubMed]
  81. Riddler, S.A.; Smit, E.; Cole, S.R.; Li, R.; Chmiel, J.S.; Dobs, A.; Palella, F.; Visscher, B.; Evans, R.; Kingsley, L.A. Impact of HIV infection and HAART on serum lipids in men. JAMA 2003, 289, 2978–2982. [Google Scholar] [CrossRef]
  82. Semu, H.; Zack, R.M.; Liu, E.; Hertzmark, E.; Spiegelman, D.; Sztam, K.; Hawkins, C.; Chalamila, G.; Muya, A.; Siril, H. Prevalence and Risk Factors for Overweight and Obesity among HIV-Infected Adults in Dar es Salaam, Tanzania. J. Int. Assoc. Provid. AIDS Care 2016, 15, 512–521. [Google Scholar] [CrossRef]
  83. Freitas, P.; Carvalho, D.; Souto, S.; Sarmento, A.; Medina, J.L. Lipodystrophy: The Metabolic Link of HIV Infection with Insulin-Resistance Syndrome. In Current Perspectives in HIV Infection; IntechOpen: London, UK, 2013. [Google Scholar] [CrossRef]
  84. Capeau, J. From lipodystrophy and insulin resistance to metabolic syndrome: HIV infection, treatment and aging. Curr. Opin. HIV AIDS 2007, 2, 247–252. [Google Scholar] [CrossRef]
  85. Boccara, F. Cardiovascular complications and atherosclerotic manifestations in the HIV-infected population: Type, incidence and associated risk factors. AIDS 2008, 22 (Suppl. S3), S19–S26. [Google Scholar] [CrossRef]
  86. Triant, V.A.; Lee, H.; Hadigan, C.; Grinspoon, S.K. Increased Acute Myocardial Infarction Rates and Cardiovascular Risk Factors among Patients with Human Immunodeficiency Virus Disease. J. Clin. Endocrinol. Metab. 2007, 92, 2506–2512. [Google Scholar] [CrossRef]
  87. Dolan, S.E.; Frontera, W.; Librizzi, J.; Ljungquist, K.; Juan, S.; Dorman, R.; Cole, M.E.; Kanter, J.R.; Grinspoon, S. Effects of a Supervised Home-Based Aerobic and Progressive Resistance Training Regimen in Women Infected with Human Immunodeficiency Virus. Arch. Intern. Med. 2006, 166, 1225. [Google Scholar] [CrossRef] [PubMed]
  88. Nguyen, N.T.P.; Tran, B.X.; Hwang, L.Y.; Markham, C.M.; Swartz, M.D.; Vidrine, J.I.; Phan, H.T.T.; Latkin, C.A.; Vidrine, D.J. Motivation to quit smoking among HIV-positive smokers in Vietnam. BMC Public Health 2015, 15, 326. [Google Scholar] [CrossRef]
  89. Al-Mansoori, L.; Al-Jaber, H.; Prince, M.S.; Elrayess, M.A. Role of Inflammatory Cytokines, Growth Factors and Adipokines in Adipogenesis and Insulin Resistance. Inflammation 2022, 45, 31–44. [Google Scholar] [CrossRef]
  90. Freitas, P.; Carvalho, D.; Santos, A.C.; Madureira, A.J.; Martinez, E.; Pereira, J.; Sarmento, A.; Medina, J.M. Adipokines, hormones related to body composition, and insulin resistance in HIV fat redistribution syndrome. BMC Infect. Dis. 2014, 14, 347. [Google Scholar] [CrossRef] [PubMed]
  91. Guerreiro, V.A.; Carvalho, D.; Freitas, P. Obesity, Adipose Tissue, and Inflammation Answered in Questions. J. Obes. 2022, 2022, 2252516. [Google Scholar] [CrossRef] [PubMed]
  92. Laroche, M.; Pécourneau, V.; Blain, H.; Breuil, V.; Chapurlat, R.; Cortet, B.; Sutter, B.; Degboe, Y.; The GRIO scientific committee. Osteoporosis and ischemic cardiovascular disease. Jt. Bone Spine 2017, 84, 427–432. [Google Scholar] [CrossRef]
  93. Shieh, A.; Greendale, G.A.; Cauley, J.A.; Srikanthan, P.; Karlamangla, A.S. Longitudinal associations of insulin resistance with change in bone mineral density in midlife women. JCI Insight 2022, 7, e162085. [Google Scholar] [CrossRef]
  94. Liu, X.; Chen, F.; Liu, L.; Zhang, Q. Prevalence of osteoporosis in patients with diabetes mellitus: A systematic review and meta-analysis of observational studies. BMC Endocr. Disord. 2023, 23, 1. [Google Scholar] [CrossRef] [PubMed]
  95. Hu, Z.; Yang, K.; Hu, Z.; Li, M.; Wei, H.; Tang, Z.; Chen, B.; Su, C.; Cai, D.; Xu, J. Determining the association between hypertension and bone metabolism markers in osteoporotic patients. Medicine 2021, 100, e26276. [Google Scholar] [CrossRef]
  96. Ahn, S.H.; Seo, D.H.; Kim, S.H.; Nam, M.S.; Hong, S. The relationship between fatty liver index and bone mineral density in Koreans: KNHANES 2010–2011. Osteoporos. Int. 2018, 29, 181–190. [Google Scholar] [CrossRef]
  97. Sowers, M.; Zheng, H.; Tomey, K.; Karvonen-Gutierrez, C.; Jannausch, M.; Li, X.; Yosef, M.; Symons, J. Changes in Body Composition in Women over Six Years at Midlife: Ovarian and Chronological Aging. J. Clin. Endocrinol. Metab. 2007, 92, 895–901. [Google Scholar] [CrossRef]
  98. Guthrie, J.R.; Dennerstein, L.; Taffe, J.R.; Ebeling, P.R.; Randolph, J.F.; Burger, H.G.; Wark, J.D. Central abdominal fat and endogenous hormones during the menopausal transition. Fertil. Steril. 2003, 79, 1335–1340. [Google Scholar] [CrossRef]
  99. Finkelstein, J.L.; Gala, P.; Rochford, R.; Glesby, M.J.; Mehta, S. HIV/AIDS and lipodystrophy: Implications for clinical management in resource-limited settings. J. Int. AIDS Soc. 2015, 18, 19033. [Google Scholar] [CrossRef]
  100. Tien, P.C.; Benson, C.; Zolopa, A.R.; Sidney, S.; Osmond, D.; Grunfeld, C. Study of Fat Redistribution and Metabolic Change in HIV Infection (FRAM): Methods, design and sample characteristics. Am. J. Epidemiol. J. 2006, 163, 860–869. [Google Scholar] [CrossRef] [PubMed]
  101. Currier, J.; Scherzer, R.; Bacchetti, P.; Heymsfield, S.; Lee, D.; Sidney, S.; Tien, P.C.; The Fat Redistribution and Metabolic Changes in HIV Infection Study Investigators. Regional Adipose Tissue and Lipid and Lipoprotein Levels in HIV-Infected Women. JAIDS J. Acquir. Immune Defic. Syndr. 2008, 48, 35–43. [Google Scholar] [CrossRef] [PubMed]
  102. Ghayomzadeh, M.; Earnest, C.P.; Hackett, D.; SeyedAlinaghi, S.; Navalta, J.W.; Gholami, M.; Hosseini, N.; Mohraz, M.; Voltarelli, F. Combination of resistance and aerobic exercise for six months improves bone mass and physical function in HIV infected individuals: A randomized controlled trial. Scand. J. Med. Sci. Sports 2021, 31, 720–732. [Google Scholar] [CrossRef] [PubMed]
  103. Hu, J.; Hu, J.; Han, D. Causal relationships between gut microbiota, plasma metabolites, and HIV infection: Insights from Mendelian randomization and mediation analysis. Virol. J. 2024, 21, 204. [Google Scholar] [CrossRef]
  104. Xu, Z.; He, P.; Xian, J.; Lu, W.; Shu, J.; Luo, W.; Gan, C.; Ke, R.; Xia, J.; Han, Z.; et al. Association between Nonalcoholic Fatty Liver Disease and Bone Mineral Density in HIV-Infected Patients Receiving Long-term TDF-Based Antiretroviral Therapy. Curr. HIV Res. 2021, 19, 40–46. [Google Scholar] [CrossRef]
  105. Dolan, S.E.; Hadigan, C.; Killilea, K.M.; Sullivan, M.P.; Hemphill, L.; Lees, R.S.; Schoenfeld, D.; Grinspoon, S. Increased Cardiovascular Disease Risk Indices in HIV-Infected Women. JAIDS J. Acquir. Immune Defic. Syndr. 2005, 39, 44–54. [Google Scholar] [CrossRef]
  106. Voulgaridou, G.; Papadopoulou, S.K.; Detopoulou, P.; Tsoumana, D.; Giaginis, C.; Kondyli, F.S.; Lymperaki, E.; Prtisa, A. Vitamin D and Calcium in Osteoporosis, and the Role of Bone Turnover Markers: A Narrative Review of Recent Data from RCTs. Diseases 2023, 11, 29. [Google Scholar] [CrossRef]
  107. BMJ Best Practice. BMJ.com. Vitamin D Deficiency-Symptoms, Diagnosis and Treatment. 2019. Available online: https://bestpractice.bmj.com/topics/en-gb/641 (accessed on 17 August 2024).
  108. Faber, J.; Bech, A.; van Bentum, P.; Gisolf, J.; Hassing, R.J.; de Boer, H. Long-Term Impact of Calcium and Vitamin D Supplementation on Bone Density in HIV+ Patients with Documented Deficiencies. AIDS Res. Hum. Retroviruses 2020, 36, 58–64. [Google Scholar] [CrossRef]
  109. Overton, E.T.; Chan, E.S.; Brown, T.T.; Tebas, P.; McComsey, G.A.; Melbourne, K.M.; Napoli, A.; Hardin, W.R.; Ribaudo, H.J.; Yin, M.T. Vitamin D and Calcium Attenuate Bone Loss With Antiretroviral Therapy Initiation. Ann. Intern. Med. 2015, 162, 815–824. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, Y.; Huang, X.; Wu, Y.; Li, A.; Tian, Y.; Ren, M.; Li, Z.; Zhang, T.; Wu, H.; Wang, W. Increased Risk of Vitamin D Deficiency Among HIV-Infected Individuals: A Systematic Review and Meta-Analysis. Front. Nutr. 2021, 8, 722032. [Google Scholar] [CrossRef]
  111. Adeyemi, O.M.; Agniel, D.; French, A.L.; Tien, P.C.; Weber, K.; Glesby, M.J.; Villacres, M.C.; Sharma, A.; Merenstein, D.; Golub, E.T.; et al. Vitamin D Deficiency in HIV-Infected and HIV-Uninfected Women in the United States. JAIDS J. Acquir. Immune Defic. Syndr. 2011, 57, 197–204. [Google Scholar] [CrossRef] [PubMed]
  112. Bi, X.; Tey, S.L.; Leong, C.; Quek, R.; Henry, C.J. Prevalence of Vitamin D Deficiency in Singapore: Its Implications to Cardiovascular Risk Factors. PLoS ONE 2016, 11, e0147616. [Google Scholar] [CrossRef] [PubMed]
  113. Chailurkit, L.O.; Aekplakorn, W.; Ongphiphadhanakul, B. Regional variation and determinants of vitamin D status in sunshine-abundant Thailand. BMC Public Health 2011, 11, 853. [Google Scholar] [CrossRef]
  114. Binkley, N.; Novotny, R.; Krueger, D.; Kawahara, T.; Daida, Y.G.; Lensmeyer, G.; Hollis, B.W.; Drezner, M.K. Low Vitamin D Status despite Abundant Sun Exposure. J. Clin. Endocrinol. Metab. 2007, 92, 2130–2135. [Google Scholar] [CrossRef]
  115. Dave, J.A.; Cohen, K.; Micklesfield, L.K.; Maartens, G.; Levitt, N.S. Antiretroviral Therapy, Especially Efavirenz, Is Associated with Low Bone Mineral Density in HIV-Infected South Africans. PLoS ONE 2015, 10, e0144286. [Google Scholar] [CrossRef]
  116. Dzavakwa, N.V.; Chisenga, M.; McHugh, G.; Filteau, S.; Gregson, C.L.; Kasonka, L.; Kranzer, K.; Mabuda, H.B.; Mujuru, H.; Redzo, N.; et al. Update: Vitamin D3 and calcium carbonate supplementation for adolescents with HIV to reduce musculoskeletal morbidity and immunopathology (VITALITY trial): Study protocol for a randomised placebo-controlled trial. Trials 2024, 25, 499. [Google Scholar] [CrossRef]
  117. Hsieh, E.; Yin, M.T. Continued Interest and Controversy: Vitamin D in HIV. Curr. HIV/AIDS Rep. 2018, 15, 199–211. [Google Scholar] [CrossRef] [PubMed]
  118. Das, S.; Bopitya, S.; Chowdhury, A.R.; Das, A.; Taha, H. Effect of Low Dose Oral Vitamin-D and Calcium Replacement in HIV Patients. Recent Pat. Antiinfect. Drug Discov. 2016, 11, 59–67. [Google Scholar] [CrossRef]
  119. Gebru, T.H.; Mekonen, H.H.; Kiros, K.G. Undernutrition and associated factors among adult HIV/AIDS patients receiving antiretroviral therapy in eastern zone of Tigray, Northern Ethiopia: A cross-sectional study. Arch. Public Health 2020, 78, 100. [Google Scholar] [CrossRef]
  120. Wadhwa, S.; Finn, T.R.; Kister, K.; Matsumura, S.; Levit, M.; Cantos, A.; Shah, J.; Bohn, B.; Lalla, E.; Grbic, J.T.; et al. Postmenopausal women with HIV have increased tooth loss. BMC Oral Health 2024, 24, 52. [Google Scholar] [CrossRef]
  121. De Cravalho, L.G.; de Fátima dos Santos Teixeira, P.; Panico, A.L.B.G.; Cohen, M.V.; Pinheiro, M.F.M.C.; Barroso, P.F.; Vaisman, M. Evaluation of thyroid function and autoimmunity in HIV-infected women. Arq. Bras. Endocrinol. Metabol. 2013, 57, 450–456. [Google Scholar] [CrossRef]
  122. Carta, S.; Olivieri, A.; Sorcini, M.; Medda, E.; Fazzini, C.; Battisti, P.; Sorcini, I.; Grandolfo, M.E. Endocrine response to HIV infection. Involvement of thyroid gland. Ann. dell’Ist. Super. Sanita 2018, 29, 451–456. [Google Scholar]
  123. Zhu, S.; Pang, Y.; Xu, J.; Chen, X.; Zhang, C.; Wu, B.; Gao, J. Endocrine Regulation on Bone by Thyroid. Front. Endocrinol. 2022, 13, 873820. [Google Scholar] [CrossRef] [PubMed]
  124. Parsa, A.A.; Bhangoo, A. HIV and thyroid dysfunction. Rev. Endocr. Metab. Disord. 2013, 14, 127–131. [Google Scholar] [CrossRef] [PubMed]
  125. Ranabir, S.; Thongam, S.; Keithelakpam, S.; Singh, T.Y.; Singh, R.L.; Singh, A.M. Thyroid dysfunction in human immunodeficiency virus-infected children and its correlation with CD4 + T lymphocyte count. Indian J. Endocrinol. Metab. 2015, 19, 272–276. [Google Scholar] [CrossRef]
  126. Ji, S.; Jin, C.; Höxtermann, S.; Fuchs, W.; Xie, T.; Lu, X.; Wu, H.; Cheng, L.; Skaletz-Rorowski, A.; Brockmeyer, N.H.; et al. Prevalence and Influencing Factors of Thyroid Dysfunction in HIV-Infected Patients. BioMed Res. Int. 2016, 2016, 1–11. [Google Scholar] [CrossRef]
  127. Sinha, U.; Sengupta, N.; Mukhopadhyay, P.; Roy, K. Human immunodeficiency virus endocrinopathy. Indian J. Endocrinol. Metab. 2011, 15, 251–260. [Google Scholar] [CrossRef]
  128. Del Ghianda, S.; Tonacchera, M.; Vitti, P. Thyroid and menopause. Climacteric 2013, 17, 225–234. [Google Scholar] [CrossRef]
  129. Delitala, A.P.; Scuteri, A.; Doria, C. Thyroid Hormone Diseases and Osteoporosis. J. Clin. Med. 2020, 9, 1034. [Google Scholar] [CrossRef]
  130. Waung, J.A.; Bassett, J.D.; Williams, G.R. Thyroid hormone metabolism in skeletal development and adult bone maintenance. Trends Endocrinol. Metab. 2012, 23, 155–162. [Google Scholar] [CrossRef]
  131. Hoffmann, C.J.; Brown, T.T. Thyroid Function Abnormalities in HIV-Infected Patients. Clin. Infect. Dis. 2007, 45, 488–494. [Google Scholar] [PubMed]
  132. Micali, C.; Russotto, Y.; Celesia, B.M.; Santoro, L.; Marino, A.; Pellicanò, G.F.; Nunnari, G.; Rullo, E.V. Thyroid Diseases and Thyroid Asymptomatic Dysfunction in People Living With HIV. Infect. Dis. Rep. 2022, 14, 655–667. [Google Scholar] [CrossRef] [PubMed]
  133. McComsey, G.A.; Tebas, P.; Shane, E.; Yin, M.T.; Overton, E.T.; Huang, J.S.; Aldrovandi, G.M.; Cardoso, S.W.; Santana, J.L.; Brown, T.T. Bone Disease in HIV infection: A Practical Review and Recommendations for HIV Care Providers. Clin. Infect. Dis. 2010, 51, 937–946. [Google Scholar] [CrossRef]
  134. Tenforde, M.W.; Walker, A.S.; Gibb, D.M.; Manabe, Y.C. Rapid antiretroviral therapy initiation in low- and middle-income countries: A resource-based approach. PLoS Med. 2019, 16, e1002723. [Google Scholar] [CrossRef]
  135. Sathi, S.R.T.T.; Anastasopoulou, C. HIV-Related Endocrinopathies; StatPearls Publishing: Treasure Island, FL, USA, 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK599549/ (accessed on 11 June 2024).
  136. Rosario, P.W. Radioiodine therapy in elderly patients with subclinical hyperthyroidism due to non-voluminous nodular goiter and its effect on bone metabolism. Arq. Bras. Endocrinol. Metabol. 2013, 57, 144–147. [Google Scholar] [CrossRef] [PubMed]
  137. Platt, L.; Easterbrook, P.; Gower, E.; McDonald, B.; Sabin, K.; McGowan, C.; Yanny, I.; Razavi, H.; Vickerman, P. Prevalence and burden of HCV co-infection in people living with HIV: A global systematic review and meta-analysis. Lancet Infect. Dis. 2016, 16, 797–808. [Google Scholar] [CrossRef]
  138. Premaor, M.; Compston, J. People living with HIV and fracture risk. Osteoporos. Int. 2020, 31, 1633–1644. [Google Scholar] [CrossRef]
  139. Dong, H.V.; Cortés, Y.I.; Shiau, S.; Yin, M.T. Osteoporosis and fractures in HIV/hepatitis C virus coinfection: A systematic review and meta-analysis. AIDS 2014, 28, 2119–2131. [Google Scholar] [CrossRef]
  140. O’neill, T.J.; Rivera, L.; Struchkov, V.; Zaheen, A.; Thein, H.-H. The Effect of HIV-Hepatitis C Co-Infection on Bone Mineral Density and Fracture: A Meta-Analysis. PLoS ONE 2014, 9, e101493. [Google Scholar] [CrossRef]
  141. Lin, M.-S.; Chen, P.-H.; Wang, P.-C.; Lin, H.-S.; Huang, T.-J.; Chang, S.-T.; Chiu, W.-N.; Chen, M.-Y. Association between hepatitis C virus infection and osteoporotic fracture risk among postmenopausal women: A cross-sectional investigation in Taiwan. BMJ Open 2019, 9, e021990. [Google Scholar] [CrossRef]
  142. González-Reimers, E.; López-Prieto, J.; Pelazas-González, R.; Alemán-Valls, M.R.; José de la Vega-Prieto, M.; Jorge-Ripper, C.; Durán-Castellón, M.C.; Santolaria-Fernández, F. Serum Sclerostin in Hepatitis C Virus Infected Patients. J. Bone Metab. 2014, 21, 69–75. [Google Scholar] [CrossRef] [PubMed]
  143. Sheng, Z.; Tong, D.; Ou, Y.; Zhang, H.; Zhang, Z.; Li, S.; Zhou, J.; Zhang, J.; Liao, E. Serum sclerostin levels were positively correlated with fat mass and bone mineral density in Central South Chinese postmenopausal women. Clin. Endocrinol. 2012, 76, 797–801. [Google Scholar] [CrossRef] [PubMed]
  144. Bedimo, R.; Maalouf, N.M.; Re, V.L.I. Hepatitis C virus coinfection as a risk factor for osteoporosis and fracture. Curr. Opin. HIV AIDS 2016, 11, 285–293. [Google Scholar] [CrossRef] [PubMed]
  145. Di Carlo, P.; Siracusa, L.; Mazzola, G.; Colletti, P.; Soresi, M.; Giannitrapani, L.; Vecchi, V.L.; Montalto, G. Vitamin D and Osteoporosis in HIV/HCV Coinfected Patients: A Literature Review. Int. J. Endocrinol. 2015, 2015, 1–7. [Google Scholar] [CrossRef] [PubMed]
  146. Ofotokun, I.; Titanji, K.; Vikulina, T.; Roser-Page, S.; Yamaguchi, M.; Zayzafoon, M.; Williams, I.R.; Weitzmann, M.N. Role of T-cell reconstitution in HIV-1 antiretroviral therapy-induced bone loss. Nat. Commun. 2015, 6, 8282. [Google Scholar] [CrossRef]
  147. Brown, T.T.; Qaqish, R.B. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: A meta-analytic review. AIDS 2006, 20, 2165–2174. [Google Scholar] [CrossRef]
  148. Biver, E. Osteoporosis and HIV Infection. Calcif. Tissue Int. 2022, 110, 624–640. [Google Scholar] [CrossRef]
  149. Ahmed, M.; Mital, D.; Abubaker, N.E.; Panourgia, M.; Owles, H.; Papadaki, I.; Ahmed, M.H. Bone Health in People Living with HIV/AIDS: An Update of Where We Are and Potential Future Strategies. Microorganisms 2023, 11, 789. [Google Scholar] [CrossRef]
  150. Baranek, B.; Wang, S.; Cheung, A.M.; Mishra, S.; Tan, D.H. The Effect of Tenofovir Disoproxil Fumarate on Bone Mineral Density: A Systematic Review and Meta-Analysis. Antivir. Ther. 2020, 25, 21–32. [Google Scholar] [CrossRef]
  151. Tourret, J.; Deray, G.; Isnard-Bagnis, C. Tenofovir Effect on the Kidneys of HIV-Infected Patients. J. Am. Soc. Nephrol. 2013, 24, 1519–1527. [Google Scholar] [CrossRef]
  152. Jacobson, M.A.; Gambertoglio, J.G.; Aweeka, F.T.; Causey, D.M.; Portale, A.A. Foscarnet-Induced Hypocalcemia and Effects of Foscarnet on Calcium Metabolism. J. Clin. Endocrinol. Metab. 1991, 72, 1130–1135. [Google Scholar] [CrossRef] [PubMed]
  153. Kuehn, E.W.; Anders, H.J.; Bogner, J.R.; Obermaier, J.; Goebel, F.D.; Schlöndorff, D. Hypocalcaemia in HIV infection and AIDS. J. Intern. Med. 1999, 245, 69–73. [Google Scholar] [CrossRef] [PubMed]
  154. Kim, S.J.; Peluso, M.J.; Wang, Y.; Bikle, D.; Shoback, D.; Kim, S. Rapid onset of hypercalcemia from high-grade lymphoma in the setting of HIV-related immune reconstitution inflammatory syndrome. Bone Rep. 2018, 10, 100194. [Google Scholar] [CrossRef] [PubMed]
  155. Starup-Linde, J.; Rosendahl, S.B.; Storgaard, M.; Langdahl, B. Management of Osteoporosis in Patients Living With HIV—A Systematic Review and Meta-analysis. Am. J. Ther. 2020, 83, 1–8. [Google Scholar] [CrossRef]
  156. Han, W.M.; Wattanachanya, L.; Apornpong, T.; Jantrapakde, J.; Avihingsanon, A.; Kerr, S.J.; Teeratakulpisarn, N.; Jadwattanakul, T.; Chaiwatanarat, T.; Buranasupkajorn, P.; et al. Bone mineral density changes among people living with HIV who have started with TDF-containing regimen: A five-year prospective study. PLoS ONE 2020, 15, e0230368. [Google Scholar] [CrossRef]
  157. Olali, A.Z.; Carpenter, K.A.; Myers, M.; Sharma, A.; Yin, M.T.; Al-Harthi, L.; Ross, R.D. Bone Quality in Relation to HIV and Antiretroviral Drugs. Curr. HIV/AIDS Rep. 2022, 19, 312–327. [Google Scholar] [CrossRef]
  158. Hoy, J. Bone Disease in HIV: Recommendations for Screening and Management in the Older Patient. Drugs Aging 2015, 32, 549–558. [Google Scholar] [CrossRef]
  159. Mansueto, P.; Seidita, A.; Vitale, G.; Gangemi, S.; Iaria, C.; Cascio, A. Vitamin D Deficiency in HIV Infection: Not Only a Bone Disorder. BioMed Res. Int. 2015, 2015, 735615. [Google Scholar] [CrossRef]
  160. Gedmintas, L.; Wright, E.A.; Losina, E.; Katz, J.N.; Solomon, D.H. Comparative risk of fracture in men and women with HIV. J. Clin. Endocrinol. Metab. 2014, 99, 486–490. [Google Scholar] [CrossRef]
  161. Bhatta, D.N.; Subedi, A.; Sharma, N. Tobacco smoking and alcohol drinking among HIV infected people using antiretroviral therapy. Tob. Induc. Dis. 2018, 16, 16. [Google Scholar] [CrossRef]
  162. Godos, J.; Giampieri, F.; Chisari, E.; Micek, A.; Paladino, N.; Forbes-Hernández, T.Y.; Quiles, J.L.; Battino, M.; La Vignera, S.; Musumeci, G.; et al. Alcohol Consumption, Bone Mineral Density, and Risk of Osteoporotic Fractures: A Dose–Response Meta-Analysis. Int. J. Environ. Res. Public Health 2022, 19, 1515. [Google Scholar] [CrossRef] [PubMed]
  163. Trevisan, C.; Alessi, A.; Girotti, G.; Zanforlini, B.M.; Bertocco, A.; Mazzochin, M.; Zoccarato, F.; Piovesan, F.; Dianin, M.; Giannini, S.; et al. The impact of smoking on bone metabolism, bone mineral density and vertebral fractures in postmenopausal women. J. Clin. Densitom. 2020, 23, 381–389. [Google Scholar] [CrossRef] [PubMed]
  164. Ellis, C.; Kruger, H.S.; Viljoen, M.; Dave, J.A.; Kruger, M.C. Factors Associated with Bone Mineral Density and Bone Resorption Markers in Postmenopausal HIV-Infected Women on Antiretroviral Therapy: A Prospective Cohort Study. Nutrients 2021, 13, 2090. [Google Scholar] [CrossRef] [PubMed]
  165. Ivers, L.C.; Cullen, K.A.; Freedberg, K.A.; Block, S.; Coates, J.; Webb, P.; Mayer, K.H. HIV/AIDS, Undernutrition, and Food Insecurity. Clin. Infect. Dis. 2009, 49, 1096–1102. [Google Scholar] [CrossRef]
  166. Takarinda, K.C.; Mutasa-Apollo, T.; Madzima, B.; Nkomo, B.; Chigumira, A.; Banda, M.; Muti, M.; Harries, A.D.; Mugurungi, O. Malnutrition status and associated factors among HIV-positive patients enrolled in ART clinics in Zimbabwe. BMC Nutr. 2017, 3, 1–11. [Google Scholar] [CrossRef]
  167. Cusano, N.E. Skeletal Effects of Smoking. Curr. Osteoporos. Rep. 2015, 13, 302–309. [Google Scholar] [CrossRef]
  168. Morgan, S.L.; Prater, G.L. Quality in dual-energy X-ray absorptiometry scans. Bone 2017, 104, 13–28. [Google Scholar] [CrossRef]
  169. Yin, M.T.; Shiau, S.; Rimland, D.; Gibert, C.L.; Bedimo, R.J.; Rodriguez-Barradas, M.C.; Harwood, K.; Aschheim, J.B.; Justice, A.C.; Womack, J.A.C. Fracture Prediction with Modified-FRAX in Older HIV-Infected and Uninfected Men. Am. J. Ther. 2016, 72, 513–520. [Google Scholar] [CrossRef] [PubMed]
  170. Vizcarra, P.; Gallego, J.; Vivancos, M.J.; Sifuentes, W.A.; Llop, M.; Casado, J.L. Evaluation of the fracture risk assessment tool for determining bone disease and the impact of secondary causes of osteoporosis in people living with HIV. HIV Clin. Trials 2020, 21, 63–71. [Google Scholar] [CrossRef]
  171. McGee, D.M.; Cotter, A.G. HIV and fracture: Risk, assessment and intervention. HIV Med. 2024, 25, 511–528. [Google Scholar] [CrossRef]
  172. Mazzitelli, M.; Isabel, P.B.; Muramatsu, T.; Chirwa, M.; Mandalia, S.; Moyle, G.; Marta, B.; Milinkovic, A. FRAX assessment in people ageing with HIV. HIV Med. 2021, 23, 103–108. [Google Scholar] [CrossRef] [PubMed]
  173. Liao, E.Y.; Wu, X.P.; Liao, H.J.; Zhang, H.; Peng, J. Effects of skeletal size of the lumbar spine on areal bone density, volumetric bone density, and the diagnosis of osteoporosis in postmenopausal women in China. J. Bone Miner. Metab. 2004, 22, 270–277. [Google Scholar] [CrossRef]
  174. Prior, J.; Burdge, D.; Maan, E.; Milner, R.; Hankins, C.; Klein, M.; Walmsley, S. Fragility fractures and bone mineral density in HIV positive women: A case-control population-based study. Osteoporos. Int. 2007, 18, 1345–1353. [Google Scholar] [CrossRef] [PubMed]
  175. Yin, M.T.; McMahon, D.J.; Ferris, D.C.; Zhang, C.A.; Shu, A.; Staron, R.; Colon, I.; Laurence, J.; Dobkin, J.F.; Hammer, S.M.; et al. Low Bone Mass and High Bone Turnover in Postmenopausal Human Immunodeficiency Virus-Infected Women. J. Clin. Endocrinol. Metab. 2010, 95, 620–629. [Google Scholar] [CrossRef] [PubMed]
  176. Long, G.; Liu, C.; Liang, T.; Zhang, Z.; Qin, Z.; Zhan, X. Predictors of osteoporotic fracture in postmenopausal women: A meta-analysis. J. Orthop. Surg. Res. 2023, 18, 1–10. [Google Scholar] [CrossRef] [PubMed]
  177. Pramukti, I.; Lindayani, L.; Chen, Y.-C.; Yeh, C.-Y.; Tai, T.-W.; Fetzer, S.; Ko, N.-Y. Bone fracture among people living with HIV: A systematic review and meta-regression of prevalence, incidence, and risk factors. PLoS ONE 2020, 15, e0233501. [Google Scholar] [CrossRef] [PubMed]
  178. Aidsmap. A Double Whammy: The Impact of HIV and Menopause on Bone Mineral Density. 2021. Available online: https://www.aidsmap.com/news/nov-2021/double-whammy-impact-hiv-and-menopause-bone-mineral-density (accessed on 6 September 2024).
  179. Wattanachanya, L.; Sunthornyothin, S.; Apornpong, T.; Lwin, H.M.S.; Kerr, S.; Gatechompol, S.; Han, W.M.; Wichiansan, T.; Siwamongsatham, S.; Chattranukulchai, P.; et al. Bone mineral density among virologically suppressed Asians older than 50 years old living with and without HIV: A cross-sectional study. PLoS ONE 2022, 17, e0277231. [Google Scholar] [CrossRef]
  180. Dravid, A.; Kulkarni, M.; Borkar, A.; Dhande, S. Prevalence of low bone mineral density among HIV patients on long-term suppressive antiretroviral therapy in resource limited setting of western India. J. Int. AIDS Soc. 2014, 17, 19567. [Google Scholar] [CrossRef]
  181. Madanhire, T.; Goedecke, J.H.; Ward, K.A.; Jaff, N.; Crowther, N.J.; Norris, S.; Ferrand, R.A.; Rehman, A.M.; Micklesfield, L.K.; Gregson, C.L. The Impact of Human Immunodeficiency Virus and Menopause on Bone Mineral Density: A Longitudinal Study of Urban-Dwelling South African Women. J. Bone Miner. Res. 2020, 38, 619–630. [Google Scholar] [CrossRef]
  182. Paruk, F.; Tsabasvi, M.; Kalla, A.A. Osteoporosis in Africa—Where are we now. Clin. Rheumatol. 2021, 40, 3419–3428. [Google Scholar] [CrossRef]
  183. Johansson, H.; Dela, S.S.; Cassim, B.; Paruk, F.; Brown, S.L.; Conradie, M.; Harvey, N.C.; Jordaan, J.D.; Kalla, A.A.; Liu, E.; et al. FRAX-based fracture probabilities in South Africa. Arch. Osteoporos. 2021, 16, 51. [Google Scholar] [CrossRef] [PubMed]
  184. Gregson, C.L.; Msc, T.M.; Rehman, A.; Ferrand, R.A.; Cappola, A.R.; Tollman, S.; Mokoena, T.; Micklesfield, L.K.; Wade, A.N.; Fabian, J.; et al. Osteoporosis, Rather Than Sarcopenia, Is the Predominant Musculoskeletal Disease in a Rural South African Community Where Human Immunodeficiency Virus Prevalence Is High: A Cross-Sectional Study. J. Bone Miner. Res. 2020, 37, 244–255. [Google Scholar] [CrossRef] [PubMed]
  185. Ward, K.A.; Pearse, C.M.; Madanhire, T.; Wade, A.N.; Fabian, J.; Micklesfield, L.K.; Gregson, C.L. Disparities in the Prevalence of Osteoporosis and Osteopenia in Men and Women Living in Sub-Saharan Africa, the UK, and the USA. Curr. Osteoporos. Rep. 2023, 21, 360–371. [Google Scholar] [CrossRef] [PubMed]
  186. Gregson, C.L.; Cassim, B.; Micklesfield, L.K.; Lukhele, M.; Ferrand, R.A.; Ward, K.A. Fragility fractures in sub-Saharan Africa: Time to break the myth. Lancet Glob. Health 2019, 7, e26–e27. [Google Scholar] [CrossRef]
  187. Kruger, M.J.; Nell, T.A. Bone mineral density in people living with HIV: A narrative review of the literature. AIDS Res. Ther. 2017, 14, 35. [Google Scholar] [CrossRef]
  188. Hamill, M.M.; Pettifor, J.M.; Ward, K.A.; Norris, S.A.; Prentice, A. Changes in Bone Mineral Density, Body Composition, Vitamin D Status, and Mineral Metabolism in Urban HIV-Positive South African Women Over 12 Months. J. Bone Miner. Res. 2017, 32, 1615–1624. [Google Scholar] [CrossRef] [PubMed]
  189. Atiase, Y.; Quarde, A. A call to action for osteoporosis research in sub-saharan Africa. Ghana Med. J. 2020, 54, 58–67. [Google Scholar] [CrossRef]
  190. Sitati, F.C.; Gichangi, P.; Obimbo, M.M. Prevalence of osteoporosis and its associated factors among postmenopausal women in Kiambu County, Kenya: A household survey. Arch. Osteoporos. 2020, 15, 1–7. [Google Scholar] [CrossRef] [PubMed]
  191. Miedany, Y.E.; Paruk, F.; Kalla, A.; Adebajo, A.; Gaafary, M.E.; Maghraoui, A.E.; Ngandeu, M.; Dey, D.; Gadallah, N.; Elwy, M.; et al. Consensus evidence-based clinical practice guidelines for the diagnosis and treat-to-target management of osteoporosis in Africa: An initiative by the African society of bone health and metabolic bone diseases. Arch Osteoporos. 2021, 16, 176. [Google Scholar] [CrossRef]
  192. Chen, X.; Orom, H.; Hay, J.L.; Waters, E.A.; Schofield, E.; Li, Y.; Kiviniemi, M.T. Differences in Rural and Urban Health Information Access and Use. J. Rural. Health 2019, 35, 405–417. [Google Scholar] [CrossRef]
  193. Matsuzaki, M.; Pant, R.; Kulkarni, B.; Kinra, S. Comparison of Bone Mineral Density between Urban and Rural Areas: Systematic Review and Meta-Analysis. PLoS ONE 2015, 10, e0132239. [Google Scholar]
  194. Gu, W.; Rennie, K.L.; Lin, X.; Wang, Y.; Yu, Z. Differences in bone mineral status between urban and rural Chinese men and women. Bone 2007, 41, 393–399. [Google Scholar] [CrossRef] [PubMed]
  195. Meyer, H.E.; Berntsen, G.K.R.; Søgaard, A.J.; Langhammer, A.; Schei, B.; Fønnebø, V.; Forsmo, S.; Tell, G.S. Higher Bone Mineral Density in Rural Compared with Urban Dwellers: The NOREPOS Study. Am. J. Epidemiol. 2004, 160, 1039–1046. [Google Scholar] [CrossRef] [PubMed]
  196. Nicholas, A.; Alare, K.; Opeyemi, M.A.; Oluwatosin, A. The outlook of rheumatological care in Africa: Current state, challenges, and recommendation. Ann. Med. Surg. 2022, 82, 104689. [Google Scholar] [CrossRef]
  197. Syed, S.T.; Gerber, B.S.; Sharp, L.K. Traveling towards Disease: Transportation Barriers to Health Care Access. J. Community Health 2013, 38, 976–993. [Google Scholar] [CrossRef]
  198. Shangase, N.; Pence, B.; Lippman, S.A.; Dufour, M.-S.K.; Kabudula, C.W.; Gómez-Olivé, F.X.; Kahn, K.; Pettifor, A. Built Environment and HIV Linkage to Care in Rural South Africa. Community Health Equity Res. Policy 2021, 43, 133–141. [Google Scholar] [CrossRef]
  199. Mbonu, N.C.; Borne, B.v.D.; De Vries, N.K. Stigma of People with HIV/AIDS in Sub-Saharan Africa: A Literature Review. J. Trop. Med. 2009, 2009, 1–14. [Google Scholar] [CrossRef] [PubMed]
  200. Rice, W.S.; Fletcher, F.E.; Akingbade, B.; Kan, M.; Whitfield, S.; Ross, S.; Gakumo, C.A.; Ofotokun, I.; Konkle-Parker, D.J.; Cohen, M.H.; et al. Quality of care for Black and Latina women living with HIV in the U.S.: A qualitative study. Int. J. Equity Health 2020, 19, 1–13. [Google Scholar] [CrossRef]
  201. Key Barriers to Women’s Access to HIV Treatment: A Global Review|UN Women—Headquarters. Available online: https://www.unwomen.org/en/digital-library/publications/2017/12/key-barriers-to-womens-access-to-hiv-treatment (accessed on 6 September 2024).
  202. Gari, S.; Doig-Acuña, C.; Smail, T.; Malungo, J.R.; Martin-Hilber, A.; Merten, S. Access to HIV/AIDS care: A systematic review of socio-cultural determinants in low and high income countries. BMC Health Serv. Res. 2013, 13, 1–198. [Google Scholar] [CrossRef]
  203. Toth, M.; Messer, L.C.; Quinlivan, E.B. Barriers to HIV Care for Women of Color Living in the Southeastern US Are Associated with Physical Symptoms, Social Environment, and Self-Determination. AIDS Patient Care STDs 2013, 27, 613–620. [Google Scholar] [CrossRef]
  204. Pienaar, M.; van Rooyen, F.C.; Walsh, C.M. Household food security and HIV status in rural and urban communities in the Free State province, South Africa. SAHARA J 2017, 14, 118–131. [Google Scholar] [CrossRef] [PubMed]
  205. Burns, P.A.; Snow, R.C. The built environment & the impact of neighborhood characteristics on youth sexual risk behavior in Cape Town, South Africa. Health Place 2012, 18, 1088–1100. [Google Scholar] [PubMed]
  206. Ellen, I.G.; Mijanovich, T.; Dillman, K.-N. Neighborhood Effects on Health: Exploring the Links and Assessing the Evidence. J. Urban Aff. 2001, 23, 391–408. [Google Scholar] [CrossRef]
  207. Santos, M.M.D.; Kruger, P.; Mellors, S.E.; Wolvaardt, G.; van der Ryst, E. An exploratory survey measuring stigma and discrimination experienced by people living with HIV/AIDS in South Africa: The People Living with HIV Stigma Index. BMC Public Health 2014, 14, 80. [Google Scholar] [CrossRef]
  208. Kang, J.-Y.; Farkhad, B.F.; Chan, M.-P.S.; Michels, A.; Albarracin, D.; Wang, S. Spatial accessibility to HIV testing, treatment, and prevention services in Illinois and Chicago, USA. PLoS ONE 2022, 17, e0270404. [Google Scholar] [CrossRef]
  209. Kimaru, L.J.; Habila, M.A.; Mantina, N.M.; Madhivanan, P.; Connick, E.; Ernst, K.; Ehiri, J. Neighborhood characteristics and HIV treatment outcomes: A scoping review. PLoS Glob. Public Heal. 2024, 4, e0002870. [Google Scholar] [CrossRef]
  210. Schinas, G.; Schinas, I.; Ntampanlis, G.; Polyzou, E.; Gogos, C.; Akinosoglou, K. Bone Disease in HIV: Need for Early Diagnosis and Prevention. Life 2024, 14, 522. [Google Scholar] [CrossRef] [PubMed]
  211. Miszkiewicz, J.J.; Cooke, K.M. Socio-economic Determinants of Bone Health from Past to Present. Clin. Rev. Bone Miner. Metab. 2019, 17, 109–122. [Google Scholar] [CrossRef]
  212. Zhu, B.; Hu, S.; Guo, J.; Dong, Z.; Dong, Y.; Li, F. Differences in the global exposure, mortality and disability of low bone mineral density between men and women: The underestimated burden in men. BMC Public Health 2023, 23, 1–13. [Google Scholar] [CrossRef]
  213. Shen, Y.; Huang, X.; Wu, J.; Lin, X.; Zhou, X.; Zhu, Z.; Pan, X.; Xu, J.; Qiao, J.; Zhang, T.; et al. The Global Burden of Osteoporosis, Low Bone Mass, and Its Related Fracture in 204 Countries and Territories, 1990–2019. Front. Endocrinol. 2022, 13, 882241. [Google Scholar] [CrossRef]
  214. Gruszczyńska, E.; Rzeszutek, M. HIV/AIDS stigma accumulation among people living with HIV: A role of general and relative minority status. Sci. Rep. 2023, 13, 10709. [Google Scholar] [CrossRef] [PubMed]
  215. Mendez-Lopez, A.; White, T.M.; Fuster-RuizdeApodaca, M.J.; Lazarus, J.V. Prevalence and sociodemographic determinants of public stigma towards people with HIV and its impact on HIVtesting uptake: A cross-sectional study in 64 low- and middle-income countries. HIV Med. 2024, 25, 83–94. [Google Scholar] [CrossRef] [PubMed]
  216. Lin, H.; Halvorsen, K.; Win, M.T.; Yancey, M.; Rbil, N.; Chatterjee, A.; Jivanelli, B.; Khormaee, S. Clinical spine care partnerships between high-income countries and low-and-middle-income countries: A scoping review. PLoS ONE 2023, 18, e0287355. [Google Scholar] [CrossRef]
  217. Erdélyi, A.; Pálfi, E.; Tűű, L.; Nas, K.; Szűcs, Z.; Török, M.; Jakab, A.; Várbíró, S. The Importance of Nutrition in Menopause and Perimenopause—A Review. Nutrients 2023, 16, 27. [Google Scholar] [CrossRef]
  218. Whitcomb, B.W.; Purdue-Smithe, A.C.; Szegda, K.L.; Boutot, M.E.; Hankinson, S.E.; Manson, J.E.; Rosner, B.; Willett, W.C.; Eliassen, A.H.; Bertone-Johnson, E.R. Cigarette Smoking and Risk of Early Natural Menopause. Am. J. Epidemiol. 2017, 187, 696–704. [Google Scholar] [CrossRef]
  219. Biver, E.; Calmy, A.; Aubry-Rozier, B.; Birkhauser, M.; Bischoff-Ferrari, H.A.; Ferrari, S.; Frey, D.; Kressig, R.W.; Lamy, O.; Lippuner, K.; et al. Diagnosis, prevention, and treatment of bone fragility in people living with HIV: A position statement from the Swiss Association against Osteoporosis. Osteoporos. Int. 2019, 30, 1125–1135. [Google Scholar] [CrossRef] [PubMed]
  220. Kebaetse, M.; Nkhwa, S.; Mogodi, M.; Masunge, J.; Gureja, Y.P.; Ramabu, M.; Mmopelwa, T.; Sharif, I.; Orford, A.; Harvey, N.C.; et al. A country-specific FRAX model for Botswana. Arch. Osteoporos. 2021, 16, 90. [Google Scholar] [CrossRef]
  221. Kumbale, C.M.; Voit, E.O. Toward Personalized Medicine for HIV/AIDS. J. AIDS HIV Treat. 2021, 3, 37–41. [Google Scholar]
  222. Lei, J.J.H.; Pereira, B.; Moyle, G.; Boffito, M.; Milinkovic, A. The benefits of tenofovir discontinuation with or without bisphosphonate therapy in osteoporotic people living with HIV. HIV Med. 2021, 22, 816–823. [Google Scholar] [CrossRef]
  223. Finnerty, F.; Walker-Bone, K.; Tariq, S. Osteoporosis in postmenopausal women living with HIV. Maturitas 2016, 95, 50–54. [Google Scholar] [CrossRef]
Figure 1. Diagram detailing the identification of studies via databases included in this narrative review.
Figure 1. Diagram detailing the identification of studies via databases included in this narrative review.
Pathogens 13 00811 g001
Figure 2. The direct and indirect impact of HIV on physiological systems leads to bone loss. Abbreviations used in the figure: BMD: Bone Mineral Density, NAFLD: Non-Alcoholic Fatty Liver Disease, HCV: Hepatitis C Virus, ARD: Antiretroviral Drugs.
Figure 2. The direct and indirect impact of HIV on physiological systems leads to bone loss. Abbreviations used in the figure: BMD: Bone Mineral Density, NAFLD: Non-Alcoholic Fatty Liver Disease, HCV: Hepatitis C Virus, ARD: Antiretroviral Drugs.
Pathogens 13 00811 g002
Table 1. Locations of different types of fractures in post-menopausal women with HIV.
Table 1. Locations of different types of fractures in post-menopausal women with HIV.
Study TypeFracture SiteFindings
Cross-sectional [42]Lumbar Spine, Femoral Neck, Total HipWLHIV had a 6.3% greater incidence of at least one vertebral fracture. WLHIV had lower femoral neck BMD (p = 0.012) and lower total hip BMD (p = 0.041). 22/104 WLHIV had osteoporosis. However, they were not found to be on any treatment.
Population-based cohort [43]Hip41.9% of the population aged 65 and above was female. HIV status was not associated with increased hip fracture risk: HR 1.02, 95% CI 0.78–1.34 p > 0.05.
Systematic review and meta-analysis [44]VertebralPLHIV had an odds ratio of vertebral fractures of 2.3 (95% CI 1.37, 3.85, I2 98.2% p < 0.00001) when compared to non-infected individuals.
Population-based cohort [45]HipOlder patients with HIV were more likely to experience a hip fracture: HR 2.11 [1.05–4.22], p = 0.035).
Population-based cohort [12]Hip, VertebralFRAX-estimated risk for hip and major osteoporotic fractures was statistically higher in PLWH (p < 0.001).
Cross-sectional [23]RadiusPost-menopausal WLHIV had a greater risk of lower BMD and risk of fracture at the proximal and distal radius (p = 0.048)
Table 2. Comparison of Antiretroviral Drug Classes and Their Impact on Bone Mineral Density (BMD) in Patients with HIV.
Table 2. Comparison of Antiretroviral Drug Classes and Their Impact on Bone Mineral Density (BMD) in Patients with HIV.
ART ClassKey DrugsMechanism of Bone LossBMD Impact FindingsSource(s)
Nucleoside Reverse Transcriptase Inhibitors (NRTIs)TDF (Tenofovir Disoproxil Fumarate), TAF (Tenofovir Alafenamide)
-
TDF linked to renal phosphate wasting, causing decreased calcium absorption and direct suppression of bone formation.
-
TDF leads to significant BMD loss, particularly in the spine and hip.
-
Switching from TDF to TAF results in BMD recovery due to reduced renal and bone toxicity.
[156]
Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs)Efavirenz (EFV), Nevirapine
-
Efavirenz reduces vitamin D levels by inducing liver enzymes, impairing calcium absorption, and leading to bone demineralization.
-
Efavirenz use is associated with bone loss, especially when combined with TDF.
-
Vitamin D supplementation can mitigate some of the bone loss associated with EFV.
[156,157]
Protease Inhibitors (PIs)Atazanavir, Darunavir
-
PIs may indirectly activate osteoclasts through increased inflammation and disrupt calcium metabolism.
-
PIs cause moderate BMD reductions, but less severe than TDF.
-
When combined with TAF, PIs show less impact on bone health than when combined with TDF.
[156,157]
Integrase Strand Transfer Inhibitors (INSTIs)Dolutegravir (DTG), Raltegravir (RAL), Elvitegravir (EVG)
-
INSTIs generally have a neutral or positive effect on BMD, with no significant negative impact on bone health.
-
Meta-analyses demonstrate better BMD preservation with INSTI-based regimens, especially when combined with TAF.
[156,157]
Combination TherapyTDF/EFV, TAF/DTG
-
TDF/EFV combinations lead to greater bone loss due to both TDF-induced phosphate wasting and EFV-induced vitamin D deficiency.
-
Switching from TDF/EFV to TAF/DTG results in significant improvements in BMD, making it a preferred switch for patients with osteoporosis or at risk.
[156,157]
Table 3. A Summary of Modifiable Risk Factors Leading to Bone Loss in Post-Menopausal WLHIV.
Table 3. A Summary of Modifiable Risk Factors Leading to Bone Loss in Post-Menopausal WLHIV.
Risk FactorDescription
Knowledge, attitude, and practice Ignorance and misconception about menopause. There is a need to increase knowledge and attitude and practice of WLHIV with regard to how to deal with menopause symptoms and protect themselves from risk of fractures [164].
Compliance with medication Ensure patients have adequate compliance with medication. Aim to discuss compliance at every health check-up or point of contact [164].
Nutrition Poor nutritional intake has been demonstrated in WLHIV, leading to vitamin D, calcium, magnesium, and phosphorus deficiencies [164]. Nutritional deficiencies in WLHIV are caused by malabsorption due to HIV and ART and worsened by food insecurity in low-income areas, leading to poor access to nutrient-rich diets [165,166].
Smoking Smoking increases the risk of osteoporosis and fractures and is more common among HIV-positive individuals [164,167].
Alcohol use Excessive alcohol consumption negatively impacts bone health and is a risk factor for low BMD [164,167].
Physical activity Exercise, including combined resistance and aerobic training, can slow down bone loss in individuals with HIV [164].
Obesity Obesity is a modifiable risk factor that can increase bone loss in individuals with HIV [164].
Table 4. Summary of osteoporosis and bone loss in WLHIV in sub-Saharan Africa, Thailand, Western India, and Brazil.
Table 4. Summary of osteoporosis and bone loss in WLHIV in sub-Saharan Africa, Thailand, Western India, and Brazil.
Study SourceStudy TypePopulationFindings
[23,40,179,180]Cross-sectionalPerimenopausal women in South Africa, HIV-infected, cART-naïve individuals in South Africa, Women in NorthWest province, South Africa
PLHIV who are older than 50 years of age in Thailand
PLHIV in western India who were on long-term cART vs. cART-naïve
Brazilian post-menopausal WLHIV on cART living in the Amazon
  • In black South African women, menopause is associated with lower BMD at the distal radius and lower cortical density at the proximal radius; 2.8% of virologically suppressed Black females had osteoporosis [18].
  • In Thai WLHIV, age and lower BMI were significantly associated with lower BMD at the lumbar spine, total hip, and femoral neck [35].
  • In Indian WLHIV, menopause is strongly associated with low BMD (p = 0.002) [155]
  • In Brazilian WLHIV, age of menopause onset is significantly earlier (p < 0.001). FRAX score was higher in WLHIV (p < 0.001). Lower T scores in lumbar spine and femoral neck following menopause onset was also seen in Brazilian WLHIV [156].
  • Older HIV-positive women with low educational status are at higher risk [156].
[181]Longitudinal (5 years)450 women aged 40–60 in Soweto, South Africa
  • Significant bone loss during menopause in women with and without HIV [157].
  • HIV-infected women had greater bone loss after adjustments [157].
[182,183,184,185,186,187,188]ReviewPost-menopausal women in South Africa, rural South Africa, sub-Saharan Africa, resource-limited settings in South Africa, African nations using cART, various African regions and ethnic groups (mainly in the Gambia, Nigeria, Kenya, and Cameroon)
  • Ethnic-specific FRAX models for South Africa available for use in HIV clinics. DEXA is the gold standard for BMD but is limited despite need for routine assessment in HIV clinics. Limited access to diagnostic and treatment options [164,165].
  • Osteoporosis more common than sarcopenia. Higher bone loss in older women with HIV. Key risk factors: HIV, malnutrition, ‘inflammaging’[166].
  • Limited access to diagnostic and treatment options [167].
  • Fracture rates expected to rise due to HIV and cART [158,162].
  • Younger individuals (<60 years) more affected [158].
  • Heel quantitative ultrasound is a cost-effective alternative [159].
  • Tenofovir Alafenamide Fumarate (TAF) is more bone-sparing than TDF but expensive [160].
  • Need for awareness of BMD decreases with TDF [160].
  • Osteoporosis and fracture rates vary across Africa [160].
  • Low awareness among population and health authorities [160].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jamshaid, M.; Heidari, A.; Hassan, A.; Mital, D.; Pearce, O.; Panourgia, M.; Ahmed, M.H. Bone Loss and Fractures in Post-Menopausal Women Living with HIV: A Narrative Review. Pathogens 2024, 13, 811. https://doi.org/10.3390/pathogens13090811

AMA Style

Jamshaid M, Heidari A, Hassan A, Mital D, Pearce O, Panourgia M, Ahmed MH. Bone Loss and Fractures in Post-Menopausal Women Living with HIV: A Narrative Review. Pathogens. 2024; 13(9):811. https://doi.org/10.3390/pathogens13090811

Chicago/Turabian Style

Jamshaid, Maryam, Amirmohammad Heidari, Ahmed Hassan, Dushyant Mital, Oliver Pearce, Maria Panourgia, and Mohamed H. Ahmed. 2024. "Bone Loss and Fractures in Post-Menopausal Women Living with HIV: A Narrative Review" Pathogens 13, no. 9: 811. https://doi.org/10.3390/pathogens13090811

APA Style

Jamshaid, M., Heidari, A., Hassan, A., Mital, D., Pearce, O., Panourgia, M., & Ahmed, M. H. (2024). Bone Loss and Fractures in Post-Menopausal Women Living with HIV: A Narrative Review. Pathogens, 13(9), 811. https://doi.org/10.3390/pathogens13090811

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop