Abstract
Purpose of the Review
The purpose of this Review was to summarize the evidence on the associations among estrogen status, cellular senescence, the gut microbiome and osteoporosis.
Recent Findings
Indicate that osteoporosis is a global public health problem that impacts individuals and society. In postmenopausal women, a decrease in estrogen levels is associated with a decrease in gut microbial diversity and richness, as well as increased permeability of the gut barrier, which allows for low-grade inflammation. The direct effects of estrogen status on the association between bone and the gut microbiome were observed in untreated and treated ovariectomized women. In addition to the direct effects of estrogens on bone remodeling, estrogen therapy could reduce the risk of postmenopausal osteoporosis by preventing increased gut epithelial permeability, bacterial translocation and inflammaging. However, in studies comparing the gut microbiota of older women, there were no changes at the phylum level, suggesting that age-related comorbidities may have a greater impact on changes in the gut microbiota than menopausal status does.
Summary
Estrogens modify bone health not only by directly influencing bone remodeling, but also indirectly by influencing the gut microbiota, gut barrier function and the resulting changes in immune system reactivity.
Keywords: Osteoporosis, Microbiota, Estrogen, Inflammation, Leaky gut, Ovariectomy, Aging
Introduction
Several factors contribute to the development of primary osteoporosis, such as aging, estrogen deficiency, low physical activity, poor nutritional status, changes in immune function, inflammatory conditions, and genetics. Aging is the most important risk factor for osteoporosis and various other age-related diseases. The loss and deterioration of bone mass progress with age from the fourth decade onward [1]. Hormonal changes play an important role in this process. These changes begin many months before the end of the menstrual cycle and can last for several years. The severity and duration of bone and hormonal changes vary from individual to individual and are influenced by genetics and exposure to various factors throughout life (exposome). Given that the population is aging, the consequences of osteoporosis and fractures are increasingly significant medical and economic problems [2]. Most fractures occur in women who do not have osteoporosis according to bone densitometry [3]. Therefore, maintaining a sufficient quantity and quality of bone mass before fractures occur is important. In this review, we aimed to emphasize that the decrease in bone mass and quality in women with increasing age and estrogen deficiency is associated with the persistence of chronic low-grade inflammation in the bones and the gut (Fig. 1). Suppressing this inflammation could be a way to reduce the risk of osteoporosis in clinical practice.
Fig. 1.
Aging and increasing bone loss are linked via dysbiosis of the intestinal microbiota, a leaky gut, low-grade inflammation and immune system senescence. SASP—senescence-associated secretory phenotype. Created with BioRender.com
Bone Aging is Associated with Estrogens
Cellular senescence is an irreversible arrest of the cell cycle that prevents apoptosis and is associated with chronic low-grade inflammation. Senescent cells produce a mixture of proinflammatory cytokines, chemokines and extracellular matrix-degrading proteins, which are referred to as the senescence-associated secretory phenotype (SASP). The SASP induces the aging of neighboring cells in an autocrine and paracrine manner, leading to chronic inflammation and the accumulation of senescent cells [4]. Senescent bone cells accumulate DNA damage, stop proliferating and actively resist apoptosis, thereby impairing their function. With age-associated thymic involution and a decline in bone marrow, the proportion of naive T and B lymphocytes decreases, and the number of autoreactive T cells increases [5–7], which may induce a proinflammatory SASP phenotype [8]. Myeloid cells and senescent osteocytes primarily develop a proinflammatory phenotype in bone [9]. The SASPs contribute to the senescence of immune cells, which share stromal progenitor cells and are subjected to the effects of coregulatory factors (RANKL, TNF-α, and IFN-γ) along with bone cells in the bone marrow. The senescence of bone marrow mesenchymal stem cells and their differentiation into adipocytes, as well as the suppression of bone formation by osteoblasts, are also induced by senescent neutrophils and monocyte-macrophages [10]. Interactions between inflammation-inducing factors and imunosenescence-inducing factors contribute to chronic inflammation; these interactions are characterized by increases in the concentrations of proinflammatory cytokines (e.g., IL-6) and acute-phase proteins [11].
Osteocytes, which are terminally differentiated cells that arise from osteoblasts, play a crucial role in bone aging [12, 13]. With increasing age, bone remodeling and bone microdamage repair are impaired by the loss of osteocytes and changes in the lacunar and canalicular networks of osteocytes [14]. Some SASPs produced by senescent osteocytes induce an increase in osteoclastic bone resorption in a paracrine manner, whereas others trigger the apoptosis of osteoblasts and osteocytes [9, 15]. Wei et al. demonstrated that the level of estrogen is negatively correlated with osteocyte and osteoblast senescence, mainly by regulating p53 protein levels. In mice, accelerated degradation of p53 after ovariectomy suppressed the expression of senescence markers and thus promoted the restoration of bone mass [16]. The removal of senescent osteocytes prevents age-related bone loss by altering the balance of bone remodeling by osteoclasts and osteoblasts [17]. In menopausal women treated with oral conjugated equine estrogen and transdermal 17β-estradiol, the serum levels of several markers of cell senescence are lower than those in women treated with a placebo [18]. Therefore, aging accelerates bone mass loss by reducing bone cell viability through the promotion of inflammation and the loss of cell protection by estrogens.
The Gut Microbiota is Associated with Aging
The human gut microbiota comprises a complex ecosystem of bacteria, archaea, fungi and viruses that cover all parts of the digestive tract. However, even in the digestive tract, the composition and function of these consortia differ at each site, with the microbiota in the large intestine or stool being the largest and best studied. Alterations in the composition and function of the microbiota are associated with a variety of inflammatory, autoimmune, metabolic and neoplastic diseases [19]. This “dysbiosis” is usually characterized by reduced microbial diversity, the excessive growth of potentially pathogenic commensals (pathobionts), the loss of commensals or a combination of these factors [20]. However, the link between diseases and their specific microbial signatures must be interpreted with caution. While this definition emphasizes the pathological feature of dysbiosis, shifts in the microbial community may be a consequence of pathology and represent an important positive mechanism of adaptation to the diseased state. Targeting the microbes associated with dysbiosis could thus undermine this compensatory mechanism. Age-related dysbiosis has been proposed as an additional feature of aging that is mechanistically linked to chronic inflammation and immune senescence [21]. However, whether this is a loss of adaptability to changing conditions or the presence or absence of key microbes, whether this is a cause or a consequence of adaptation, or even what is low microbial diversity, remains controversial and may differ for each disease [22].
Owing to the abundance of energy, stable temperature, low oxygen content and many other factors that create a specific niche in the human gut, the vast majority of all bacteria in the typical colonic microbiota of healthy people belong to four phyla: Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria [23]. However, changes in several external factors, such as diet, have a profound impact on these consortia even in healthy individuals. Switching from a plant-based to an animal-based diet triggers a shift from Alistipes, Bilophila and Bacteroides to Roseburia, Eubacterium rectale and Ruminococcus bromii [24], and the abundance of 15% of all microbes fluctuates daily, which is simply due to the alternation between eating and fasting [25]. This shaping of the gut microbiota can influence the ability of the immune system to induce inflammation, which in turn affects bone health [26]. However, while the gut microbiota and the formation of short-chain fatty acids change with diet, multiple studies analyzing specific diets (e.g., the Mediterranean diet) often do not show clear or consistent effects on the gut microbiota [27]. This discrepancy could be due to the considerable interindividual variability in the composition of the microbiota caused by other variables, such as the age of the participants [28] or the degree of urbanization of their environment [29].
The gut microbiota changes with age. In the first 3 years of life, microbial consortia rapidly develop as the infant’s gut adapts to the environment and changes in food sources. Even if the main components of the gut microbiota are already firmly established at this time, the gut microbiota will continue to change in older people. This development is associated with other factors associated with aging, such as low-grade inflammation, immunosenescence or loss of gut barrier integrity and bone mass. In both women and men, the function of intestinal epithelial cells and gut barrier integrity deteriorate with age, changes in hormonal status, and other environmental factors [30]. The gut microbiota of older people also differs from that of younger people, but these changes occur gradually and without a chronological threshold [28]. The microbiota of older people appears to be less stable than that of younger people [31]. From the fourth decade of life onward, the relative abundance of Actinobacteria, Bifidobacteria and several members of the phylum Firmicutes with anti-inflammatory properties, including Clostridium cluster IV and Clostridium cluster XIVa, decreases [32, 33], and the abundance of Proteobacteria and Bacteroidetes increases [34–37]. The combination of probiotics and prebiotics, which leads to anti-inflammatory tuning of immunity, may help control inflammation in elderly individuals and promote healthy aging (see [38]).
In people older than 100 years, osteoporosis develops much more slowly in the oldest age group than in younger centenarians or in the control population [39]. Moreover, people older than 105 years have a greater gut microbiota diversity and different gut microbiota representations (higher representation of the genera Akkermansia and Bifidobacterium and the family Christensenellaceae) than does the general population of centenarians [40, 41]. These data were obtained mainly by comparing cohorts of people of different ages. This cross-sectional study design cannot distinguish whether these microbial characteristics are a cause or consequence of longevity or whether they are caused only by a specific condition at the time of colonization and are not related to longevity. Lactobacillus amylovorus in the gut is associated with an increased likelihood of longevity, whereas F. nucleatum is negatively associated with longevity [40]. However, this microbial signature is associated with decreased body adiposity [42] and a decreased risk of colorectal cancer [43], which could influence longevity. Nevertheless, these associations are still part of the emerging science and studies finding this microbial signature are criticized for their technical, methodological or design shortcomings and because only some isolates of the associated bacteria can drive the disease [44, 45].
The Gut Microbiota is Associated with Estrogen Status
The intestinal microbiota modulates estrogen levels [46], and estrogens also influence the intestinal microbiota [47]. In humans, genes from more than 60 genera of gut bacteria, collectively referred to as the estrobolome, can metabolize estrogens. These include β-glucuronidases, which deconjugate estrogens into biologically active forms and enable their reuptake into the bloodstream [48]. However, the gut microbiota also converts polycyclic aromatic hydrocarbons into estrogenic compounds, suggesting a mechanism by which these pollutants disrupt the endocrine system [49]. The estrogen status also significantly impacts gut motility, which indirectly alters the gut microbiota community [50] and gut microbiota can influence gut motility via the release of metabolites or end products of bacterial fermentation [51]. Therefore, sex steroids alter the microbiota and the microbiota influences its metabolism, creating important communication pathways between the host and microbes. These pathways utilize hypothalamic–pituitary–adrenal activity and autonomic nervous system function. While both estrogens and androgens are influenced by the microbiota, each is impacted differently, which has important implications for sex- and age-related differences in disease susceptibility [52, 53].
The production of sex hormones is an important factor for interindividual differences in the diversity of the gut microbiota. The absence of sex hormones reduces the expression of tight junction proteins, which affects the structure of tight junctions and impairs the function of the intestinal barrier [54, 55]. Therefore, both dysbiosis and intestinal barrier failure can be accelerated by the absence of sex hormones [55, 56]. The relationship between sex hormones and the gut microbiota has been shown in a number of studies in rodents, allowing for better control of interindividual variability, and demonstrating an increase in the relative abundance of Firmicutes and Proteobacteria, with decreases in Bacteroidetes [57–61]. Ovariectomy in rats significantly reduced species richness and significantly shifted β diversity as early as 4 weeks after ovariectomy. This shift is caused by a significantly increased proportion of several bacterial species (Helicobacter rodentium, Lachnospiraceae 10–1, Lachnospiraceae A4 and Lactobacillus reuteri) and shows marked variability between individual rats. Estrogen supplementation not only counteracts the adverse conditions resulting from ovariectomy but also shifts the microbiota toward its normal composition [62]. These studies showed that the gut microbiota responds to sex hormone levels. However, the translation of these results to humans might be complicated by key differences related to the hormonal system (estrous vs. menstrual cycle), the gut (much shorter, faster passage), the diet (well controlled in laboratory rodents) and the variability of the microbiota (clean and controlled environment in laboratory animal facilities).
The results of studies comparing the fecal microbiota in different groups of estrogen-deficient women are summarized in Table 1. In summary, sex hormone levels influence several physiological states, but their influence on the gut microbiota in postmenopausal women is less clear [63, 64]. In some populations, a decrease in bone mass is associated with a particular microbial pattern in which some representatives of Ruminococcus are associated with low bone mineral density (BMD) (Table 1) [65, 66]. However, changes at the phylum level have not been reported in any studies [67, 68], suggesting that sex and comorbidities have a greater impact on changes in the gut microbiota than menopausal status does. Obesity appears to be one of the most important confounding factors in these studies [69], as controlling for age and obesity may fundamentally alter the patterns of microbiota associated with postmenopausal status [70].
Table 1.
Association of taxonomic changes in the gut microbiota with sex hormone deficiency* or reduced bone mass* The differences are toward group marked with * (↑ = increased in * group; (p) = phylum, (c) = class, (o) = order, (f) = family, (g) = genus)
α diversity | Specific microbes | Group definition | Reference |
---|---|---|---|
No difference |
↑ Firmicutes (p), ↑ Prevotella (g), ↑ Bilophila (g), ↓ Actinobacteria (p), ↓ Parabacteroides (g), ↓ Lachnospira (g), ↓ Roseburia (g), |
Premenopausal (mean age 46 yrs) vs. postmenopausal* women (mean age 56 yrs) | [67] |
↓ Shannon index, ↓ Genus richness |
↑ Bacteroidetes (g) ↑ Tolumonas (g) ↓ Firmicutes (p) ↓Roseburia (g) |
Premenopausal (mean age 53 yrs) vs. postmenopausal* women (mean age 54 yrs) |
[71] |
No difference |
↑ Pasteurellaceae (f), ↑ Comamonadaceae (f), ↑ Lentisphaerae (f) ↓ Acholeplasmataceae (f), ↓ Enterococcaceae (f), ↓ Micrococcales (f), ↓ Christensenellaceae (f), ↓ Ruminococcus sp CAG:379, ↓ Clostridium neonatale |
Premenopausal (mean age 42 yrs) vs. postmenopausal* women (mean age 59 yrs) |
[70] |
↓ Observed Species, ↓ Shannon index, (No association between E2 and bacterial community structure) |
↑ Lactobacillales (o), ↑ Coriobacteriales (o), ↑ Parabacteroides (g), ↑ Lactobacillus (g) ↓Bacteroides massiliensis, ↓ Lachnospira pectinoschiza, ↓Bacteroides coprocola ↓ Blautia (g) |
Postmenopausal women (mean age 58 yrs) with normal bone mass vs. women with postmenopausal osteoporosis and lower estrogen level* | [72] |
Not analyzed |
↑ Rikenellaceae (f) ↓ Bacteroides (g) |
Postmenopausal women with higher* vs. lower fracture risk and lower* vs. higher BMD, respectively (mean age 63 yrs) | [73] |
No difference |
↑ Bacteroidetes (p), ↑ Bacteriodes (g), ↑ Bifidobacterium (g), ↑ Megamonas (g), ↑ Prevotella (g), ↑ Dorea (g), ↑ Sutterella (g), ↑ Butyricimonas (g) ↓ Firmicutes (p), ↓ Faecalibacterium (g), ↓ Clostridium (g), ↓ Coprococcus (g), ↓ Roseburia (g), ↓ Ruminococcus (g), |
Women with premature ovarian insufficiency (mean age 35 yrs) * vs. healthy women (mean age 34 yrs) | [74] |
No difference |
↑ Clostridium XLVa (g), ↑ Coprococcus (g), ↑ Lactobacillus (g), ↑ Eggerthella (g), ↑ Parabacteroides (g), ↑ Flavonifractor (g) ↓ Veillonella (g), ↑ Raoultella (g) |
Men and women with osteoporosis* (mean age 70 yrs) vs. controls (mean age 68 yrs) | [75] |
No difference in Shannon or Simpson diversity indices |
↑ Bacteroides (g), ↑ Betaproteobacteria (c), ↑ Bacteroides stercoris, ↑ Adlercreutzia (g), ↓ Clostridia (c), ↓ Methanobacteriaceae (f), ↓ Peptostreptococcaceae (f), ↓ Turicibacter (g), ↓ Romboutsia (g), ↓ unclassified Coriobacteriia |
Postmenopausal women with normal bone mass (mean age 63 yrs) vs. postmenopausal women with osteoporosis (mean age 65 yrs) * | [76] |
No differences in Chao1 and Shannon diversity indices |
↑ Lactobacillus (g), ↑ Ruminococcus (g), ↑ Bacteroides (g) ↓ Blautia (g), ↓ Alistipes (g) |
Meta-analysis, healthy men and women vs. people with osteoporosis* | [65] |
↓ Shannon index |
↑ Bacteroides sp.Ga6A1, Prevotella marshii, Sutterella wadsworthensis, ↓ Escherichia coli, Veillonella seminalis, Parabacteroides johnsonii, [Clostridium] lactatifermentans, Oscillibacter sp.KLE1745 |
Premenopausal (mean age 40 yrs) vs. postmenopausal (mean age 53 yrs) * women | [77] |
↓ Shannon index, no differences in observed ASVs and Faith’s PD |
E1: ↑ Eubacterium ventriosum group (g), ↑ Mollicutes.RF39 (f) E2: ↓ Burkholderiaceae (f), ↑ Intestinibacter (g), ↑ unknown Clostridiales (g), |
Low levels* of estrone (E1) and estradiol (E2) in postmenopausal women (mean age 58 yrs) | [78] |
No difference |
↑ Fusobacteria (p), ↑ Bacilli (c), ↑ Erysipelotrichia (c), ↑ unidentified Clostridiales (g), ↑ Clostridium disporicum, ↑ Lactobacillus salivarius ↓ Ruminococcaceae (f), ↓ Bacteroides eggerthii |
Postmenopausal women with normal bone mass (mean age 57 yrs) vs. postmenopausal women with postmenopausal osteoporosis (mean age 59 yrs)* | [79] |
↓ Faith PD decrease after ≤ 12 months | Minimal changes, ↓ Lactococcus lactis | Paired samples of women (mean age 48 yrs) before vs. after ovariectomy* | [69] |
↓ Observed Species, ↓ Shannon index | ↓ Alistipes (g), ↓Collinsella (g), ↓ Erysipelotrichia (c), ↓ Clostridia (c) | Low levels* of estrogens in HIV + postmenopausal women (mean age 58 yrs) | [80] |
Leaky Gut and Inflammation Link the Gut Microbiota and Bone Health
Studies in both men and women have shown a significant correlation between the richness and diversity of the gut microbiota and bone mass [81, 82]. This correlation may not be solely due to low levels of sex hormones. Although estrogen deficiency drives bone loss in conventional mice, it is not sufficient to increase bone resorption or trabecular bone loss in germ-free mice [54]. Several specific microbial shifts are associated with low bone mass (Table 1), suggesting that there is a pathogenetic link between the gut microbiota and bone mass and quality.
Certain microbes and their products can improve intestinal barrier function and reduce inflammation. Oral administration of Prevotella histicola to ovariectomized mice increased the expression of tight junction proteins (zonula occludens-1 and occludin), prevented TNF-α overproduction, and halted bone loss induced by estrogen deficiency [83]. The use of probiotics also dampens chronic low-grade inflammation, although this effect may depend on the suitability of specific probiotics for the population [84]. However, improvements in bone metabolism and the prevention of bone loss could also be achieved by certain microbial products. Butyrate, a short-chain fatty acid, has anti-inflammatory effects by inhibiting NF-κB activation in intestinal epithelial cells [85] and by interacting with immune cells through specific receptors [86]. Owing to this immunomodulatory effect, a reduction in butyrate-producing bacteria, such as members of the Firmicutes phylum (e.g., Roseburia spp., and Faecalibacterium prausnitzii), could lead to a deterioration of intestinal barrier function and even intestinal inflammation. By producing butyrate, members of the genus Lachnospiraceae (phylum Firmicutes), whose abundance is positively correlated with bone mass, improve the function of the intestinal barrier and prevent the transfer of bacterial lipopolysaccharides and chronic low-grade inflammation [87].
Metabolites from the gut microbiota establish a link between the microbiota and bone health in various ways. They promote bone growth and remodeling by inducing the production of insulin-like growth factor 1 (IGF-1) [88], although some specific microbes can promote bone growth through IGF-1 signaling via the innate immune receptor NOD2 in the gut [89]. SCFAs also induce the production of the gut hormones glucagon-like peptide-1 (GLP-1), GLP-2 and peptide YY from the enteroendocrine L cells of the gut. GLP-1 and GLP-2 promote bone formation, whereas the peptide YY promotes bone resorption [90, 91]. While the microbiota can promote both mechanisms, the composition of microbial signals and differences in microbial production dynamics play key roles in selecting the dominant mechanism [92].
Vitamin D is another pleiotropic factor that influences bone health directly [93], but also indirectly through modulation of the gut microbiota, gut barrier function, immune response, intestinal transcellular and paracellular transport, gut lumen pH, and short-chain fatty acid production [94]. Studies in mice suggest that the gut microbiota upregulates vitamin D receptor (VDR) through fermentation products. Loss of the VDR gene (by genetic manipulation in mice) or a VDR gene polymorphism in humans explains a significant part of the variability in the gut microbiota. Vitamin D also contributes to the maintenance of gut barrier integrity by ensuring adequate levels of antimicrobial peptides and mucus production and by strengthening intercellular junctions [95]. The effects of vitamin D may be further modulated by estrogen deficiency. Estradiol is positively associated with intestinal calcium absorption via an estrogen receptor [96, 97]. In addition, estrogen upregulates the expression of vitamin D receptor (VDR) in the rat duodenal mucosa and increases the responsiveness to endogenous 1,25(OH)2D [98]. Thus, vitamin D deficiency could lead to a leaky gut and inappropriate stimulation of the immune system, creating proinflammatory conditions. Vitamin D also regulates the adaptive immune system by suppressing Th1/Th17 cells and promoting Treg cells [99, 100], which could further exacerbate the inflammatory response in patients with vitamin D deficiency.
In summary, leaky gut and chronic low-grade inflammation could be common denominators linking dysbiosis, sex hormone loss and the aging of bone cells. Disruption of the intestinal barrier causes the immune system to enter a proinflammatory state, during which the cytokines and immune cells can spread throughout the body and affect bone mass and quality.
Bone and Gut Microbiota After Menopause
Fluctuating sex hormone levels during the menstrual cycle as well as the maintenance of constant levels of estrogen and progesterone by oral contraceptives might impact the diversity and differences in the abundance of several bacterial taxa [101, 102]. Accelerated bone loss after menopause is difficult to separate from aging, and ovariectomy may not be the perfect model for evaluating menopause. While both ovariectomy and menopause lead to estrogen deficiency and are associated with accelerated bone loss, the dynamics of estrogen loss differ. Ovariectomy does not reflect the dynamics of immune responses during the physiological menopausal transition. In women, ovariectomy is associated with thymic hypertrophy [6], whereas physiologically, thymic involution ends by the fourth decade [5]. In older individuals, the vast majority of lymphocytes reside in gut-associated lymphoid tissues [103, 104]. Conversely, thymectomy significantly suppresses T-cell lymphopoiesis and bone resorption in mice after ovariectomy [105]. Nevertheless, ovariectomy is a model that can be used to evaluate the impact of estrogen deficiency alone on the relationship between the gut microbiota and bone mass. Estrogen deficiency after ovariectomy is associated with further increases in RANKL production, and osteoclastogenesis, prolongation of the lifespan of osteoclasts, increased osteoclastic bone resorption, and accelerated bone loss [106]. Osteocytes, B cells, and differentiated Th17 TNF-α+ cells enhance the effect of RANKL through the production of IL-17 and TNF-α [6] [107–111]. Decreased secretion of osteoprotegerin by osteocytes, as well as B cells and dendritic cells [112], prevents the interaction between RANKL and RANK and contributes to increased osteoclastogenesis and prolongation of the lifespan of osteoclasts [107]. Interestingly, RANKL is also expressed by epithelial cells throughout the intestine. The administration of exogenous RANKL to RANKL-deficient mice has demonstrated a critical role for RANKL in the differentiation of RANK-expressing enterocytes into microfold (M) cells, which are important in the induction of specific mucosal immune responses in Peyer’s patches [113–116]. There is an immunological connection between the bone marrow and the digestive tract. Ovariectomy in mice leads to increased gut permeability and subsequently increased bacterial translocation. Experimental studies have clearly demonstrated a link between intestinal dysbiosis and bone loss [61] and an association between intestinal barrier dysfunction in the context of gut dysbiosis and a reduction in bone mass [117]. These mechanisms potentiate one other, as the dysbiotic gut microbiota interacts more strongly with the immune system when the gut barrier is disturbed, resulting in chronic low-grade inflammation.
This inflammation leads to increased local production of IFN-γ and TNF-α, which exacerbates gut barrier dysfunction by decreasing the expression of tight junction proteins in epithelial and endothelial cells [118, 119]. TNF-α increases the number of Th17 cells (CD4+ IL-17A+ T lymphocytes) in the intestine and directs the migration of Th17 cells from the intestine to the bone marrow by upregulating CCL20 [120]. Translocated gut microbes, proinflammatory microbial components (e.g., lipopolysaccharides), cytokines and stimulated immune cells can enter the systemic circulation and induce changes in distant organs, including bone loss. The antigen-independent production of IL-17 and TNF-α by memory T cells during the loss of estrogen production is driven by the simultaneous overproduction of IL-7 and IL-15 by dendritic cells in the bone marrow [121]. IL-17 stimulates the release of RANKL from osteoblasts and osteocytes [122] and upregulates RANK expression, thereby increasing the osteoclastogenic activity of RANKL. IL-17, RANKL or TNF-α blocking antibodies prevent the deterioration of trabecular microarchitecture in ovariectomized mice [123]. By blocking the exit of T lymphocytes from the intestine into the bloodstream and their entry into the bone marrow, both the expansion of T cells in the bone marrow and trabecular bone loss are prevented [120]. Blocking T-cell migration or treatment with TNF inhibitors [108] could therefore be an alternative to prevent bone resorption in estrogen-deficient postmenopausal women. These findings show the importance of immune system activation for bone resorption in the absence of estrogen.
The dynamics of changes in bone mass and the fecal microbiota after ovariectomy in women are not as rapid as those reported in the experimental studies mentioned above. According to a prospective controlled study of women ovariectomized at their reproductive age, the fecal microbiota diversity did not change during the first six months after surgery compared to that before surgery, whereas the bone remodeling marker values increased significantly, and the lumbar spine BMD decreased significantly. The decrease in lumbar spine BMD continued over the following year, whereas the rate of bone remodeling remained elevated. However, after ovariectomy, it took 18 months for the gut microbiota to significantly change [69]. As the direct effects of estradiol deficiency on the bone marrow can be observed in women in the first months after ovariectomy, further studies should investigate whether and to what extent the migration of T lymphocytes from the intestinal lymphoid tissue contributes to bone marrow expansion.
Menopausal Hormone Therapy and the Gut Microbiota
The presence of estrogen receptors (ERα, ERβ, and the G-protein-coupled receptor GPER) and androgen receptors in the intestinal epithelium [124] explains the effects of hormone therapy on the diversity of the gut microbiota and intestinal barrier function in experimental studies [54, 59, 125, 126]. In postmenopausal women treated with estrogens, the abundance of Proteobacteria and Bacteroidetes in the duodenum reached premenopausal levels [127]. In women, one year of daily low-dose estradiol treatment prevented the decrease in the diversity of the intestinal microbiota typical of women not receiving hormone treatment [69]. However, further studies are needed to evaluate the associations of estrogen levels with changes in the gut microbiota and the aging of intestinal cells [128–131]. As hormone therapy in women after physiological and artificial menopause [69, 127] alters the state of the gut microbiota, further studies are needed to verify the extent to which the decline in bone mass after several years in postmenopausal women is influenced by the migration of T lymphocytes from the lymphoid tissue of the gut. The differentiation of Th17 cells in humans can be triggered by two dozen nonpathogenic intestinal bacteria [132]. Blocking the migration of T lymphocytes from the intestine to the bone marrow could be a therapeutic alternative for preventing postmenopausal bone loss. An inhibitor of the sphingosine-1-phosphate receptor stops the transport of lymphocytes from Peyer's patches and mesenteric lymph nodes without affecting the function of lymphocytes and reduces the number of Th17 cells in the bone marrow [133].
Questions, Problems, Controversies and the Way Forward
There is a constant bidirectional interaction among the gut microbiota, sex hormones and immune system and bone health can be influenced by these interactions. Key mechanisms include dysbiosis, an increase in gut permeability and chronic low-grade inflammation. However, despite a wealth of valuable and intriguing findings, the search for microbial biomarkers has not yet identified any universal microbial taxa that could be used to target or predict age-related bone loss. The gut microbiota of estrogen-deficient women is usually less diverse than that of healthy individuals and often has a similar pattern of enrichment (e.g., A. muciniphila) or depletion (e.g., several members of the genus Clostridium) across multiple studies. The significance and relevance of these changes are difficult to assess owing to the variability of study results and the presence of numerous age-related comorbidities. In addition, only some microbes of a particular species may influence disease progression [45]. Therefore, multiomics integration of the available microbial community data may be needed to identify the most important microbial factors [134].
The gut microbiota adapts to the environment, and its high diversity may represent the ability of the microbiome to react to the ever-changing world. In fact, a decrease in gut microbiota diversity may signal specialization of the gut microbiota, which could improve efficiency in the less volatile environment of the industrialized world [135]. There is still a lack of knowledge on how the function of one microbe can impact another, but it is clear that the metabolic activity of the gut microbiota is much less volatile than its taxonomic composition [136]. For example, a significant increase in estrogen levels during the third trimester of pregnancy is associated with decreased phylogenetic diversity and a significant shift in the β diversity of the gut microbiota without significant changes in the microbiome's gene repertoire [137]. Thus, studies comparing microbial metabolism may lead to faster recognition of key microbial traits than studies of microbial taxonomy, especially if there are no paired samples from prospective studies available. However, for interactions with the immune system, structural components of the microbiota are still quite important, and these structural components can differ between microbial species or even between isolates and can be missed if only metabolism is studied [138].
Most related research has focused on the microbiota in feces, which is the largest and most easily accessible source of samples. However, an important interaction between the immune system and microbes occurs in the organized lymphoid tissue of the small intestine (e.g., Peyer’s patches). Thus, minor differences in microbial composition could be important because of the lower microbial load in the small intestine. The effects of sex hormones on the small intestinal microbiota have rarely been studied, and these microbes may be underrepresented in the stool, so small changes are easy to miss. Nevertheless, the duodenal microbiota responds to the loss of estrogens during menopause and to hormone treatment after menopause [127], suggesting that this topic is interesting for future research. These interactions may even occur during the "window of opportunity" in early life, making them even more challenging to study. The immune system (and other physiological functions) develops in infancy, and changes immune system reactivity may manifest as impaired immune system regulation later in life [139]. To develop new hypotheses suitable for clinical evaluation, studies must first be conducted in gnotobiotic models and carefully implemented in humans.
The gut microbiota, the estrobolome, clearly responds to sex hormones and has the ability to influence their metabolism. However, it is difficult to assess the relative importance of estrogen levels, as these levels may be specific to the microbes colonizing a particular individual and compensated for by the organism’s own feedback mechanisms. The increased absorption of estrogens facilitated by the gut microbiota may be compensated for by increased urinary excretion, thereby allowing the estrogens saved from stool to exit the organism via urine [140, 141]. Therefore, a careful description of the estrogen balance should be considered in future studies.
The effects of menopause, ovariectomy or even pregnancy are often summarized as changes in sex hormones. However, these are independent processes associated with their own list of age-related problems or lifestyle factors. Ovariectomy is perfect for analyzing the effect of estrogens on the microbiota (and bone), but may not be the perfect model for evaluating menopause. Both lead to estrogen deficiency, and both are associated with osteoporosis, but the dynamics of estrogen loss are different. Menopause is not just the loss of estrogen; postmenopausal osteoporosis is difficult to separate from aging. The extent of the inflammatory response and gut barrier failure may influence not only bone mass, but also other symptoms of menopause [142]. Obesity, leaky gut and some diseases associated with aging are known modulators of the microbiota. Therefore, the results of these studies must be interpreted with full knowledge of these confounding factors.
Conclusion
Primary osteoporosis is a global public health problem for individuals and society. The timely prevention of low-impact fractures is becoming increasingly urgent. Chronic low-grade inflammation, triggered by hormonal changes, leaky gut and cellular senescence, could represent an interesting therapeutic target. Suppressing inflammation appears to be a viable strategy for reducing the risk of osteoporosis. This strategy has been demonstrated for physical activity [143] and menopausal hormone therapy [144]. Estrogens suppress bone and immune cell senescence, decrease RANKL production, reduce osteoclastogenesis, prolong the lifespan of osteoclasts, decrease osteoclastic bone resorption, and attenuate bone loss. Interestingly, changes in the gut microbiota are associated with most steps in the pathogenesis of osteoporosis, making them prominent targets for novel therapeutic research. Some gut microbes are associated with an increase in the return of estrogens to the host, improved gut barrier function and dampened inflammation. However, deciphering the complex pathogenesis of osteoporosis and the mechanisms linking the gut microbiota to bone health is complicated by numerous age-related comorbidities.
Author Contributions
M.K. and J.J.S. wrote the main manuscript text, M.K. prepared Fig. 1 and Table 1. All authors reviewed the manuscript.
Funding
Open access publishing supported by the National Technical Library in Prague. MK is supported by the Czech Academy of Sciences under the Lumina quaeruntur fellowship No. LQ200202105 and the Ministry of Education, Youth and Sports of the Czech Republic grant Talking Microbes—understanding microbial interactions within One Health framework (CZ.02.01.01/00/22_008/0004597). JS is supported by the RVO 00023728 (Institute of Rheumatology), and Charles University Project SVV 260 523.
Data Availability
No datasets were generated or analysed during the current study.
Declarations
Competing Interests
The authors declare no competing interests.
Human and Animal Rights and Informed Consent
All the reported data from the human studies performed by the authors have been previously published and complied with all the applicable ethical standards including the Helsinki Declaration and its amendments institutional/national research committee standards and guidelines.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Kanis JA, Burlet N, Cooper C, Delmas PD, Reginster JY, Borgstrom F, et al. European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporos Int. 2008;19(4):399–428. 10.1007/s00198-008-0560-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Willers C, Norton N, Harvey NC, Jacobson T, Johansson H, Lorentzon M, et al. Osteoporosis in Europe: a compendium of country-specific reports. Arch Osteoporos. 2022;17(1):23. 10.1007/s11657-021-00969-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Trajanoska K, Schoufour JD, de Jonge EAL, Kieboom BCT, Mulder M, Stricker BH, et al. Fracture incidence and secular trends between 1989 and 2013 in a population based cohort: The Rotterdam Study. Bone. 2018;114:116–24. 10.1016/j.bone.2018.06.004. [DOI] [PubMed] [Google Scholar]
- 4.da Silva PFL, Ogrodnik M, Kucheryavenko O, Glibert J, Miwa S, Cameron K, et al. The bystander effect contributes to the accumulation of senescent cells in vivo. Aging Cell. 2019;18(1):e12848. 10.1111/acel.12848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Steinmann GG, Klaus B, Muller-Hermelink HK. The involution of the ageing human thymic epithelium is independent of puberty. A morphometric study Scand J Immunol. 1985;22(5):563–75. 10.1111/j.1365-3083.1985.tb01916.x. [DOI] [PubMed] [Google Scholar]
- 6.Adeel S, Singh K, Vydareny KH, Kumari M, Shah E, Weitzmann MN, et al. Bone loss in surgically ovariectomized premenopausal women is associated with T lymphocyte activation and thymic hypertrophy. J Investig Med. 2013;61(8):1178–83. 10.2310/JIM.0000000000000016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Thomas R, Wang W, Su DM. Contributions of Age-Related Thymic Involution to Immunosenescence and Inflammaging. Immun Ageing. 2020;17:2. 10.1186/s12979-020-0173-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pangrazzi L, Weinberger B. T cells, aging and senescence. Exp Gerontol. 2020;134:110887. 10.1016/j.exger.2020.110887. [DOI] [PubMed] [Google Scholar]
- 9.Farr JN, Fraser DG, Wang H, Jaehn K, Ogrodnik MB, Weivoda MM, et al. Identification of Senescent Cells in the Bone Microenvironment. J Bone Miner Res. 2016;31(11):1920–9. 10.1002/jbmr.2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li CJ, Xiao Y, Sun YC, He WZ, Liu L, Huang M, et al. Senescent immune cells release grancalcin to promote skeletal aging. Cell Metab. 2021;33(10):1957-73 e6. 10.1016/j.cmet.2021.08.009. [DOI] [PubMed] [Google Scholar]
- 11.Bano G, Trevisan C, Carraro S, Solmi M, Luchini C, Stubbs B, et al. Inflammation and sarcopenia: A systematic review and meta-analysis. Maturitas. 2017;96:10–5. 10.1016/j.maturitas.2016.11.006. [DOI] [PubMed] [Google Scholar]
- 12.Liu ZJ, Zhuge Y, Velazquez OC. Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem. 2009;106(6):984–91. 10.1002/jcb.22091. [DOI] [PubMed] [Google Scholar]
- 13.Mi B, Xiong Y, Knoedler S, Alfertshofer M, Panayi AC, Wang H, et al. Ageing-related bone and immunity changes: insights into the complex interplay between the skeleton and the immune system. Bone Research. 2024;12(1):42. 10.1038/s41413-024-00346-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ru JY, Wang YF. Osteocyte apoptosis: the roles and key molecular mechanisms in resorption-related bone diseases. Cell Death Dis. 2020;11(10):846. 10.1038/s41419-020-03059-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Josephson AM, Bradaschia-Correa V, Lee S, Leclerc K, Patel KS, Muinos Lopez E, et al. Age-related inflammation triggers skeletal stem/progenitor cell dysfunction. Proc Natl Acad Sci U S A. 2019;116(14):6995–7004. 10.1073/pnas.1810692116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wei Y, Fu J, Wu W, Ma P, Ren L, Wu J. Estrogen prevents cellular senescence and bone loss through Usp10-dependent p53 degradation in osteocytes and osteoblasts: the role of estrogen in bone cell senescence. Cell Tissue Res. 2021;386(2):297–308. 10.1007/s00441-021-03496-7. [DOI] [PubMed] [Google Scholar]
- 17.Farr JN, Xu M, Weivoda MM, Monroe DG, Fraser DG, Onken JL, et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med. 2017;23(9):1072–9. 10.1038/nm.4385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Faubion L, White TA, Peterson BJ, Geske JR, LeBrasseur NK, Schafer MJ, et al. Effect of menopausal hormone therapy on proteins associated with senescence and inflammation. Physiol Rep. 2020;8(16):e14535. 10.14814/phy2.14535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tlaskalova-Hogenova H, Stepankova R, Kozakova H, Hudcovic T, Vannucci L, Tuckova L, et al. The role of gut microbiota (commensal bacteria) and the mucosal barrier in the pathogenesis of inflammatory and autoimmune diseases and cancer: contribution of germ-free and gnotobiotic animal models of human diseases. Cell Mol Immunol. 2011;8(2):110–20. 10.1038/cmi.2010.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17(4):219–32. 10.1038/nri.2017.7. [DOI] [PubMed] [Google Scholar]
- 21.Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA. Chronic inflammation and the hallmarks of aging. Mol Metab. 2023;74:101755. 10.1016/j.molmet.2023.101755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kverka M, Tlaskalova-Hogenova H. Intestinal Microbiota: Facts and Fiction. Dig Dis. 2017;35(1–2):139–47. 10.1159/000449095. [DOI] [PubMed] [Google Scholar]
- 23.Bajer L, Kverka M, Kostovcik M, Macinga P, Dvorak J, Stehlikova Z, et al. Distinct gut microbiota profiles in patients with primary sclerosing cholangitis and ulcerative colitis. World J Gastroenterol. 2017;23(25):4548–58. 10.3748/wjg.v23.i25.4548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63. 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Thaiss CA, Zeevi D, Levy M, Zilberman-Schapira G, Suez J, Tengeler AC, et al. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell. 2014;159(3):514–29. 10.1016/j.cell.2014.09.048. [DOI] [PubMed] [Google Scholar]
- 26.Byberg L, Bellavia A, Larsson SC, Orsini N, Wolk A, Michaelsson K. Mediterranean Diet and Hip Fracture in Swedish Men and Women. J Bone Miner Res. 2016;31(12):2098–105. 10.1002/jbmr.2896. [DOI] [PubMed] [Google Scholar]
- 27.Kimble R, Gouinguenet P, Ashor A, Stewart C, Deighton K, Matu J, et al. Effects of a mediterranean diet on the gut microbiota and microbial metabolites: A systematic review of randomized controlled trials and observational studies. Crit Rev Food Sci Nutr. 2023;63(27):8698–719. 10.1080/10408398.2022.2057416. [DOI] [PubMed] [Google Scholar]
- 28.O’Toole PW, Jeffery IB. Gut microbiota and aging. Science. 2015;350(6265):1214–5. 10.1126/science.aac8469. [DOI] [PubMed] [Google Scholar]
- 29.Gupta VK, Paul S, Dutta C. Geography, Ethnicity or Subsistence-Specific Variations in Human Microbiome Composition and Diversity. Front Microbiol. 2017;8:1162. 10.3389/fmicb.2017.01162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Xu Y, Liu X, Liu X, Chen D, Wang M, Jiang X, et al. The Roles of the Gut Microbiota and Chronic Low-Grade Inflammation in Older Adults With Frailty. Front Cell Infect Microbiol. 2021;11:675414. 10.3389/fcimb.2021.675414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Claesson MJ, Cusack S, O’Sullivan O, Greene-Diniz R, de Weerd H, Flannery E, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4586–91. 10.1073/pnas.1000097107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Biagi E, Nylund L, Candela M, Ostan R, Bucci L, Pini E, et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE. 2010;5(5):e10667. 10.1371/journal.pone.0010667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ren J, Li H, Zeng G, Pang B, Wang Q, Wei J. Gut microbiome-mediated mechanisms in aging-related diseases: are probiotics ready for prime time? Frontiers in Pharmacology. 2023;14:1178596. 10.3389/fphar.2023.1178596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, et al. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90. 10.1186/s12866-016-0708-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Xu C, Zhu H, Qiu P. Aging progression of human gut microbiota. BMC Microbiol. 2019;19(1):236. 10.1186/s12866-019-1616-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Badal VD, Vaccariello ED, Murray ER, Yu KE, Knight R, Jeste DV, et al. The Gut Microbiome, Aging, and Longevity: A Systematic Review. Nutrients. 2020;12(12):3759. 10.3390/nu12123759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bosco N, Noti M. The aging gut microbiome and its impact on host immunity. Genes Immun. 2021;22(5):289–303. 10.1038/s41435-021-00126-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Warman DJ, Jia H, Kato H. The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging). Nutrients. 2022;14(4):747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Andersen SL, Sebastiani P, Dworkis DA, Feldman L, Perls TT. Health span approximates life span among many supercentenarians: compression of morbidity at the approximate limit of life span. J Gerontol A Biol Sci Med Sci. 2012;67(4):395–405. 10.1093/gerona/glr223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Liu X, Zou L, Nie C, Qin Y, Tong X, Wang J, et al. Mendelian randomization analyses reveal causal relationships between the human microbiome and longevity. Sci Rep. 2023;13(1):5127. 10.1038/s41598-023-31115-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Biagi E, Franceschi C, Rampelli S, Severgnini M, Ostan R, Turroni S, et al. Gut Microbiota and Extreme Longevity. Current biology : CB. 2016;26(11):1480–5. 10.1016/j.cub.2016.04.016. [DOI] [PubMed] [Google Scholar]
- 42.Omar JM, Chan Y-M, Jones ML, Prakash S, Jones PJH. Lactobacillus fermentum and Lactobacillus amylovorus as probiotics alter body adiposity and gut microflora in healthy persons. J Funct Foods. 2013;5(1):116–23. 10.1016/j.jff.2012.09.001. [Google Scholar]
- 43.Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14(2):207–15. 10.1016/j.chom.2013.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.He Y, Mujagond P, Tang W, Wu W, Zheng H, Chen X, et al. Non-nucleatum Fusobacterium species are dominant in the Southern Chinese population with distinctive correlations to host diseases compared with F. nucleatum. Gut. 2021;70(4):810–2. 10.1136/gutjnl-2020-322090. [DOI] [PubMed] [Google Scholar]
- 45.Zepeda-Rivera M, Minot SS, Bouzek H, Wu H, Blanco-Míguez A, Manghi P, et al. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature. 2024;628(8007):424–32. 10.1038/s41586-024-07182-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas. 2017;103:45–53. 10.1016/j.maturitas.2017.06.025. [DOI] [PubMed] [Google Scholar]
- 47.Chen KL, Madak-Erdogan Z. Estrogen and Microbiota Crosstalk: Should We Pay Attention? Trends Endocrinol Metab. 2016;27(11):752–5. 10.1016/j.tem.2016.08.001. [DOI] [PubMed] [Google Scholar]
- 48.Kwa M, Plottel CS, Blaser MJ, Adams S. The Intestinal Microbiome and Estrogen Receptor-Positive Female Breast Cancer. J Natl Cancer Inst. 2016;108(8):djw029. 10.1093/jnci/djw029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Van de Wiele T, Vanhaecke L, Boeckaert C, Peru K, Headley J, Verstraete W, et al. Human colon microbiota transform polycyclic aromatic hydrocarbons to estrogenic metabolites. Environ Health Perspect. 2005;113(1):6–10. 10.1289/ehp.7259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hogan AM, Collins D, Baird AW, Winter DC. Estrogen and its role in gastrointestinal health and disease. Int J Colorectal Dis. 2009;24(12):1367–75. 10.1007/s00384-009-0785-0. [DOI] [PubMed] [Google Scholar]
- 51.Waclawiková B, Codutti A, Alim K, El Aidy S. Gut microbiota-motility interregulation: insights from in vivo, ex vivo and in silico studies. Gut Microbes. 2022;14(1):1997296. 10.1080/19490976.2021.1997296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.So SY, Savidge TC. Sex-Bias in Irritable Bowel Syndrome: Linking Steroids to the Gut-Brain Axis. Front Endocrinol. 2021;12:684096. 10.3389/fendo.2021.684096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.So SY, Savidge TC. Gut feelings: the microbiota-gut-brain axis on steroids. Am J Physiol Gastrointest Liver Physiol. 2022;322(1):G1–20. 10.1152/ajpgi.00294.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Li JY, Chassaing B, Tyagi AM, Vaccaro C, Luo T, Adams J, et al. Sex steroid deficiency-associated bone loss is microbiota dependent and prevented by probiotics. J Clin Invest. 2016;126(6):2049–63. 10.1172/JCI86062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Shieh A, Epeldegui M, Karlamangla AS, Greendale GA. Gut permeability, inflammation, and bone density across the menopause transition. JCI Insight. 2020;5(2). 10.1172/jci.insight.134092. [DOI] [PMC free article] [PubMed]
- 56.Salazar AM, Aparicio R, Clark RI, Rera M, Walker DW. Intestinal barrier dysfunction: an evolutionarily conserved hallmark of aging. Dis Model Mech. 2023;16(4):dmm049969. 10.1242/dmm.049969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Benedek G, Zhang J, Nguyen H, Kent G, Seifert HA, Davin S, et al. Estrogen protection against EAE modulates the microbiota and mucosal-associated regulatory cells. J Neuroimmunol. 2017;310:51–9. 10.1016/j.jneuroim.2017.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen KLA, Zhao YC, Hieronymi K, Smith BP, Madak-Erdogan Z. Bazedoxifene and conjugated estrogen combination maintains metabolic homeostasis and benefits liver health. PLoS ONE. 2017;12(12):e0189911. 10.1371/journal.pone.0189911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, et al. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome. 2018;6(1):205. 10.1186/s40168-018-0587-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Acharya KD, Gao X, Bless EP, Chen J, Tetel MJ. Estradiol and high fat diet associate with changes in gut microbiota in female ob/ob mice. Sci Rep. 2019;9(1):20192. 10.1038/s41598-019-56723-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang N, Meng F, Ma S, Fu L. Species-level gut microbiota analysis in ovariectomized osteoporotic rats by Shallow shotgun sequencing. Gene. 2022;817:146205. 10.1016/j.gene.2022.146205. [DOI] [PubMed] [Google Scholar]
- 62.Meng Q, Ma M, Zhang W, Bi Y, Cheng P, Yu X, et al. The gut microbiota during the progression of atherosclerosis in the perimenopausal period shows specific compositional changes and significant correlations with circulating lipid metabolites. Gut Microbes. 2021;13(1):1–27. 10.1080/19490976.2021.1880220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.d’Afflitto M, Upadhyaya A, Green A, Peiris M. Association Between Sex Hormone Levels and Gut Microbiota Composition and Diversity-A Systematic Review. J Clin Gastroenterol. 2022;56(5):384–92. 10.1097/MCG.0000000000001676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yang M, Wen S, Zhang J, Peng J, Shen X, Xu L. Systematic Review and Meta-analysis: Changes of Gut Microbiota before and after Menopause. Dis Markers. 2022;2022:3767373. 10.1155/2022/3767373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Huang R, Liu P, Bai Y, Huang J, Pan R, Li H, et al. Changes in the gut microbiota of osteoporosis patients based on 16S rRNA gene sequencing: a systematic review and meta-analysis. J Zhejiang Univ Sci B. 2022;23(12):1002–13. 10.1631/jzus.B2200344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Xue Y, Wang X, Liu H, Kang J, Liang X, Yao A, et al. Assessment of the relationship between gut microbiota and bone mineral density: a two-sample Mendelian randomization study. Front Microbiol. 2024;15:1298838. 10.3389/fmicb.2024.1298838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Santos-Marcos JA, Rangel-Zuniga OA, Jimenez-Lucena R, Quintana-Navarro GM, Garcia-Carpintero S, Malagon MM, et al. Influence of gender and menopausal status on gut microbiota. Maturitas. 2018;116:43–53. 10.1016/j.maturitas.2018.07.008. [DOI] [PubMed] [Google Scholar]
- 68.Zhu J, Liao M, Yao Z, Liang W, Li Q, Liu J, et al. Breast cancer in postmenopausal women is associated with an altered gut metagenome. Microbiome. 2018;6(1):136. 10.1186/s40168-018-0515-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Jackova Z, Stepan JJ, Coufal S, Kostovcik M, Galanova N, Reiss Z, et al. Interindividual differences contribute to variation in microbiota composition more than hormonal status: A prospective study. Front Endocrinol. 2023;14:1139056. 10.3389/fendo.2023.1139056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mayneris-Perxachs J, Arnoriaga-Rodriguez M, Luque-Cordoba D, Priego-Capote F, Perez-Brocal V, Moya A, et al. Gut microbiota steroid sexual dimorphism and its impact on gonadal steroids: influences of obesity and menopausal status. Microbiome. 2020;8(1):136. 10.1186/s40168-020-00913-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhao H, Chen J, Li X, Sun Q, Qin P, Wang Q. Compositional and functional features of the female premenopausal and postmenopausal gut microbiota. FEBS Lett. 2019;593(18):2655–64. 10.1002/1873-3468.13527. [DOI] [PubMed] [Google Scholar]
- 72.He J, Xu S, Zhang B, Xiao C, Chen Z, Si F, et al. Gut microbiota and metabolite alterations associated with reduced bone mineral density or bone metabolic indexes in postmenopausal osteoporosis. Aging (Albany NY). 2020;12(9):8583–604. 10.18632/aging.103168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ozaki D, Kubota R, Maeno T, Abdelhakim M, Hitosugi N. Association between gut microbiota, bone metabolism, and fracture risk in postmenopausal Japanese women. Osteoporos Int. 2021;32(1):145–56. 10.1007/s00198-020-05728-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wu J, Zhuo Y, Liu Y, Chen Y, Ning Y, Yao J. Association between premature ovarian insufficiency and gut microbiota. BMC Pregnancy Childbirth. 2021;21(1):418. 10.1186/s12884-021-03855-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wei M, Li C, Dai Y, Zhou H, Cui Y, Zeng Y, et al. High-Throughput Absolute Quantification Sequencing Revealed Osteoporosis-Related Gut Microbiota Alterations in Han Chinese Elderly. Front Cell Infect Microbiol. 2021;11:630372. 10.3389/fcimb.2021.630372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Rettedal EA, Ilesanmi-Oyelere BL, Roy NC, Coad J, Kruger MC. The Gut Microbiome Is Altered in Postmenopausal Women With Osteoporosis and Osteopenia. JBMR Plus. 2021;5(3):e10452. 10.1002/jbm4.10452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Peters BA, Xue X, Sheira LA, Qi Q, Sharma A, Santoro N, et al. Menopause Is Associated With Immune Activation in Women With HIV. J Infect Dis. 2022;225(2):295–305. 10.1093/infdis/jiab341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wu Z, Pfeiffer RM, Byrd DA, Wan Y, Ansong D, Clegg-Lamptey JN, et al. Associations of Circulating Estrogens and Estrogen Metabolites with Fecal and Oral Microbiome in Postmenopausal Women in the Ghana Breast Health Study. Microbiol Spectr. 2023;11(4):e0157223. 10.1128/spectrum.01572-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huang D, Wang J, Zeng Y, Li Q, Wang Y. Identifying microbial signatures for patients with postmenopausal osteoporosis using gut microbiota analyses and feature selection approaches. Front Microbiol. 2023;14:1113174. 10.3389/fmicb.2023.1113174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Peters B, Hanna D, Wang Y, Weber K, Topper E, Appleton A, et al. Sex Hormones, the Stool Microbiome, and Subclinical Atherosclerosis in Women With and Without HIV. J Clin Endocrinol Metab. 2024;109(2):483–97. 10.1210/clinem/dgad510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wang J, Wang Y, Gao W, Wang B, Zhao H, Zeng Y, et al. Diversity analysis of gut microbiota in osteoporosis and osteopenia patients. PeerJ. 2017;5:e3450. 10.7717/peerj.3450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ling CW, Miao Z, Xiao ML, Zhou H, Jiang Z, Fu Y, et al. The Association of Gut Microbiota With Osteoporosis Is Mediated by Amino Acid Metabolism: Multiomics in a Large Cohort. J Clin Endocrinol Metab. 2021;106(10):e3852–64. 10.1210/clinem/dgab492. [DOI] [PubMed] [Google Scholar]
- 83.Wang Z, Chen K, Wu C, Chen J, Pan H, Liu Y, et al. An emerging role of Prevotella histicola on estrogen deficiency-induced bone loss through the gut microbiota-bone axis in postmenopausal women and in ovariectomized mice. Am J Clin Nutr. 2021;114(4):1304–13. 10.1093/ajcn/nqab194. [DOI] [PubMed] [Google Scholar]
- 84.Custodero C, Mankowski RT, Lee SA, Chen Z, Wu S, Manini TM, et al. Evidence-based nutritional and pharmacological interventions targeting chronic low-grade inflammation in middle-age and older adults: A systematic review and meta-analysis. Ageing Res Rev. 2018;46:42–59. 10.1016/j.arr.2018.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, Giardina C. The luminal short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell line. Gastroenterology. 2000;118(4):724–34. 10.1016/s0016-5085(00)70142-9. [DOI] [PubMed] [Google Scholar]
- 86.Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50. 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
- 87.Huang S, Chen J, Cui Z, Ma K, Wu D, Luo J, et al. Lachnospiraceae-derived butyrate mediates protection of high fermentable fiber against placental inflammation in gestational diabetes mellitus. Sci Adv. 2023;9(44):eadi7337. 10.1126/sciadv.adi7337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yan J, Charles JF. Gut Microbiota and IGF-1. Calcif Tissue Int. 2018;102(4):406–14. 10.1007/s00223-018-0395-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Schwarzer M, Gautam UK, Makki K, Lambert A, Brabec T, Joly A, et al. Microbe-mediated intestinal NOD2 stimulation improves linear growth of undernourished infant mice. Science. 2023;379(6634):826–33. 10.1126/science.ade9767. [DOI] [PubMed] [Google Scholar]
- 90.Zeng Y, Wu Y, Zhang Q, Xiao X. Crosstalk between glucagon-like peptide 1 and gut microbiota in metabolic diseases. mBio. 2024;15(1):e0203223. 10.1128/mbio.02032-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Liu H, Xiao H, Lin S, Zhou H, Cheng Y, Xie B, et al. Effect of gut hormones on bone metabolism and their possible mechanisms in the treatment of osteoporosis. Front Pharmacol. 2024;15:1372399. 10.3389/fphar.2024.1372399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Covasa M, Stephens RW, Toderean R, Cobuz C. Intestinal Sensing by Gut Microbiota: Targeting Gut Peptides. Front Endocrinol. 2019;10:82. 10.3389/fendo.2019.00082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Bikle DD. Vitamin D and bone. Curr Osteoporos Rep. 2012;10:151–9. 10.1007/s11914-012-0098-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Menon R, Watson SE, Thomas LN, Allred CD, Dabney A, Azcarate-Peril MA, et al. Diet complexity and estrogen receptor beta status affect the composition of the murine intestinal microbiota. Appl Environ Microbiol. 2013;79(18):5763–73. 10.1128/AEM.01182-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Yamamoto EA, Jorgensen TN. Relationships Between Vitamin D, Gut Microbiome, and Systemic Autoimmunity. Front Immunol. 2019;10:3141. 10.3389/fimmu.2019.03141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Colin EM, Van Den Bemd GJ, Van Aken M, Christakos S, De Jonge HR, Deluca HF, et al. Evidence for involvement of 17beta-estradiol in intestinal calcium absorption independent of 1,25-dihydroxyvitamin D3 level in the Rat. J Bone Miner Res. 1999;14(1):57–64. 10.1359/jbmr.1999.14.1.57. [DOI] [PubMed] [Google Scholar]
- 97.Ten Bolscher M, Netelenbos JC, Barto R, Van Buuren LM, Vandervijgh WJ. Estrogen regulation of intestinal calcium absorption in the intact and ovariectomized adult rat. J Bone Miner Res. 1999;14(7):1197–202. 10.1359/jbmr.1999.14.7.1197. [DOI] [PubMed] [Google Scholar]
- 98.Liel Y, Shany S, Smirnoff P, Schwartz B. Estrogen increases 1,25-dihydroxyvitamin D receptors expression and bioresponse in the rat duodenal mucosa. Endocrinology. 1999;140(1):280–5. 10.1210/endo.140.1.6408. [DOI] [PubMed] [Google Scholar]
- 99.Fakhoury HMA, Kvietys PR, AlKattan W, Anouti FA, Elahi MA, Karras SN, et al. Vitamin D and intestinal homeostasis: Barrier, microbiota, and immune modulation. J Steroid Biochem Mol Biol. 2020;200:105663. 10.1016/j.jsbmb.2020.105663. [DOI] [PubMed] [Google Scholar]
- 100.Fawaz L, Mrad MF, Kazan JM, Sayegh S, Akika R, Khoury SJ. Comparative effect of 25(OH)D3 and 1,25(OH)2D3 on Th17 cell differentiation. Clin Immunol. 2016;166–167:59–71. 10.1016/j.clim.2016.02.011. [DOI] [PubMed] [Google Scholar]
- 101.Schieren A, Koch S, Pecht T, Simon MC. Impact of Physiological Fluctuations of Sex Hormones During the Menstrual Cycle on Glucose Metabolism and the Gut Microbiota. Exp Clin Endocrinol Diabetes. 2024;132(5):267–78. 10.1055/a-2273-5602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Mihajlovic J, Leutner M, Hausmann B, Kohl G, Schwarz J, Röver H, et al. Combined hormonal contraceptives are associated with minor changes in composition and diversity in gut microbiota of healthy women. Environ Microbiol. 2021;23(6):3037–47. 10.1111/1462-2920.15517. [DOI] [PubMed] [Google Scholar]
- 103.Vighi G, Marcucci F, Sensi L, Di Cara G, Frati F. Allergy and the gastrointestinal system. Clin Exp Immunol. 2008;153(Suppl 1):3–6. 10.1111/j.1365-2249.2008.03713.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mörbe UM, Jørgensen PB, Fenton TM, von Burg N, Riis LB, Spencer J, et al. Human gut-associated lymphoid tissues (GALT); diversity, structure, and function. Mucosal Immunol. 2021;14(4):793–802. 10.1038/s41385-021-00389-4. [DOI] [PubMed] [Google Scholar]
- 105.Ryan MR, Shepherd R, Leavey JK, Gao Y, Grassi F, Schnell FJ, et al. An IL-7-dependent rebound in thymic T cell output contributes to the bone loss induced by estrogen deficiency. Proc Natl Acad Sci U S A. 2005;102(46):16735–40. 10.1073/pnas.0505168102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Weitzmann MN, Pacifici R. Estrogen deficiency and bone loss: an inflammatory tale. J Clin Invest. 2006;116(5):1186–94. 10.1172/JCI28550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, Riggs BL. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest. 2003;111(8):1221–30. 10.1172/JCI17215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Charatcharoenwitthaya N, Khosla S, Atkinson EJ, McCready LK, Riggs BL. Effect of blockade of TNF-alpha and interleukin-1 action on bone resorption in early postmenopausal women. J Bone Miner Res. 2007;22(5):724–9. 10.1359/jbmr.070207. [DOI] [PubMed] [Google Scholar]
- 109.Pacifici R. Role of T cells in ovariectomy induced bone loss–revisited. J Bone Miner Res. 2012;27(2):231–9. 10.1002/jbmr.1500. [DOI] [PubMed] [Google Scholar]
- 110.Ono T, Hayashi M, Sasaki F, Nakashima T. RANKL biology: bone metabolism, the immune system, and beyond. Inflamm Regen. 2020;40:2. 10.1186/s41232-019-0111-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Marahleh A, Kitaura H, Ohori F, Noguchi T, Mizoguchi I. The osteocyte and its osteoclastogenic potential. Front Endocrinol. 2023;14:1121727. 10.3389/fendo.2023.1121727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Bengtsson AK, Ryan EJ. Immune function of the decoy receptor osteoprotegerin. Crit Rev Immunol. 2002;22(3):201–15. [PubMed] [Google Scholar]
- 113.Knoop KA, Kumar N, Butler BR, Sakthivel SK, Taylor RT, Nochi T, et al. RANKL is necessary and sufficient to initiate development of antigen-sampling M cells in the intestinal epithelium. J Immunol. 2009;183(9):5738–47. 10.4049/jimmunol.0901563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Mabbott NA, Donaldson DS, Ohno H, Williams IR, Mahajan A. Microfold (M) cells: important immunosurveillance posts in the intestinal epithelium. Mucosal Immunol. 2013;6(4):666–77. 10.1038/mi.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Nakamura Y, Kimura S, Hase K. M cell-dependent antigen uptake on follicle-associated epithelium for mucosal immune surveillance. Inflamm Regen. 2018;38(1):15. 10.1186/s41232-018-0072-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kimura S, Nakamura Y, Kobayashi N, Shiroguchi K, Kawakami E, Mutoh M, et al. Osteoprotegerin-dependent M cell self-regulation balances gut infection and immunity. Nat Commun. 2020;11(1):234. 10.1038/s41467-019-13883-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wang N, Ma S, Fu L. Gut Microbiota Dysbiosis as One Cause of Osteoporosis by Impairing Intestinal Barrier Function. Calcif Tissue Int. 2022;110(2):225–35. 10.1007/s00223-021-00911-7. [DOI] [PubMed] [Google Scholar]
- 118.Naydenov NG, Baranwal S, Khan S, Feygin A, Gupta P, Ivanov AI. Novel mechanism of cytokine-induced disruption of epithelial barriers: Janus kinase and protein kinase D-dependent downregulation of junction protein expression. Tissue Barriers. 2013;1(4):e25231. 10.4161/tisb.25231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ozaki H, Ishii K, Horiuchi H, Arai H, Kawamoto T, Okawa K, et al. Cutting edge: combined treatment of TNF-alpha and IFN-gamma causes redistribution of junctional adhesion molecule in human endothelial cells. J Immunol. 1999;163(2):553–7. [PubMed] [Google Scholar]
- 120.Yu M, Pal S, Paterson CW, Li JY, Tyagi AM, Adams J, et al. Ovariectomy induces bone loss via microbial-dependent trafficking of intestinal TNF+ T cells and Th17 cells. J Clin Invest. 2021;131(4). 10.1172/JCI143137. [DOI] [PMC free article] [PubMed]
- 121.Cline-Smith A, Axelbaum A, Shashkova E, Chakraborty M, Sanford J, Panesar P, et al. Ovariectomy Activates Chronic Low-Grade Inflammation Mediated by Memory T Cells, Which Promotes Osteoporosis in Mice. J Bone Miner Res. 2020;35(6):1174–87. 10.1002/jbmr.3966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Li JY, D’Amelio P, Robinson J, Walker LD, Vaccaro C, Luo T, et al. IL-17A Is Increased in Humans with Primary Hyperparathyroidism and Mediates PTH-Induced Bone Loss in Mice. Cell Metab. 2015;22(5):799–810. 10.1016/j.cmet.2015.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Tyagi AM, Mansoori MN, Srivastava K, Khan MP, Kureel J, Dixit M, et al. Enhanced immunoprotective effects by anti-IL-17 antibody translates to improved skeletal parameters under estrogen deficiency compared with anti-RANKL and anti-TNF-alpha antibodies. J Bone Miner Res. 2014;29(9):1981–92. 10.1002/jbmr.2228. [DOI] [PubMed] [Google Scholar]
- 124.Dart DA, Waxman J, Aboagye EO, Bevan CL. Visualising androgen receptor activity in male and female mice. PLoS ONE. 2013;8(8):e71694. 10.1371/journal.pone.0071694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Wada-Hiraike O, Imamov O, Hiraike H, Hultenby K, Schwend T, Omoto Y, et al. Role of estrogen receptor beta in colonic epithelium. Proc Natl Acad Sci U S A. 2006;103(8):2959–64. 10.1073/pnas.0511271103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Peters BA, Santoro N, Kaplan RC, Qi Q. Spotlight on the Gut Microbiome in Menopause: Current Insights. Int J Women’s Health. 2022;14:1059–72. 10.2147/IJWH.S340491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Leite G, Barlow G, Parodi G, Pimentel M, Chang C, Hosseini A, et al. Duodenal microbiome changes in postmenopausal women: effects of hormone therapy and implications for cardiovascular risk. Menopause. 2022;29(3):264–75. 10.1097/GME.0000000000001917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kawano N, Koji T, Hishikawa Y, Murase K, Murata I, Kohno S. Identification and localization of estrogen receptor alpha- and beta-positive cells in adult male and female mouse intestine at various estrogen levels. Histochem Cell Biol. 2004;121(5):399–405. 10.1007/s00418-004-0644-6. [DOI] [PubMed] [Google Scholar]
- 129.Moorefield EC, Andres SF, Blue RE, Van Landeghem L, Mah AT, Santoro MA, et al. Aging effects on intestinal homeostasis associated with expansion and dysfunction of intestinal epithelial stem cells. Aging (Albany NY). 2017;9(8):1898–915. 10.18632/aging.101279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Sehl ME, Ganz PA. Potential Mechanisms of Age Acceleration Caused by Estrogen Deprivation: Do Endocrine Therapies Carry the Same Risks? JNCI Cancer Spectrum. 2018;2(3):pky035. 10.1093/jncics/pky035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Hohman LS, Osborne LC. A gut-centric view of aging: Do intestinal epithelial cells contribute to age-associated microbiota changes, inflammaging, and immunosenescence? Aging Cell. 2022;21(9):e13700. 10.1111/acel.13700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tan TG, Sefik E, Geva-Zatorsky N, Kua L, Naskar D, Teng F, et al. Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proc Natl Acad Sci U S A. 2016;113(50):E8141–50. 10.1073/pnas.1617460113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Yu M, Malik Tyagi A, Li JY, Adams J, Denning TL, Weitzmann MN, et al. PTH induces bone loss via microbial-dependent expansion of intestinal TNF(+) T cells and Th17 cells. Nat Commun. 2020;11(1):468. 10.1038/s41467-019-14148-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Chetty A, Blekhman R. Multi-omic approaches for host-microbiome data integration. Gut Microbes. 2024;16(1):2297860. 10.1080/19490976.2023.2297860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Gomez A, Petrzelkova KJ, Burns MB, Yeoman CJ, Amato KR, Vlckova K, et al. Gut Microbiome of Coexisting BaAka Pygmies and Bantu Reflects Gradients of Traditional Subsistence Patterns. Cell Rep. 2016;14(9):2142–53. 10.1016/j.celrep.2016.02.013. [DOI] [PubMed] [Google Scholar]
- 136.Human Microbiome Project C. Structure function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–14. 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Koren O, Goodrich JK, Cullender TC, Spor A, Laitinen K, Backhed HK, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell. 2012;150(3):470–80. 10.1016/j.cell.2012.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Pena JA, Li SY, Wilson PH, Thibodeau SA, Szary AJ, Versalovic J. Genotypic and phenotypic studies of murine intestinal lactobacilli: species differences in mice with and without colitis. Appl Environ Microbiol. 2004;70(1):558–68. 10.1128/AEM.70.1.558-568.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ottman N, Smidt H, de Vos WM, Belzer C. The function of our microbiota: who is out there and what do they do? Front Cell Infect Microbiol. 2012;2:104. 10.3389/fcimb.2012.00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Adlercreutz H, Pulkkinen MO, Hamalainen EK, Korpela JT. Studies on the role of intestinal bacteria in metabolism of synthetic and natural steroid hormones. J Steroid Biochem. 1984;20(1):217–29. 10.1016/0022-4731(84)90208-5. [DOI] [PubMed] [Google Scholar]
- 141.Fuhrman BJ, Feigelson HS, Flores R, Gail MH, Xu X, Ravel J, et al. Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women. J Clin Endocrinol Metab. 2014;99(12):4632–40. 10.1210/jc.2014-2222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Chen TY, Huang WY, Liu KH, Kor CT, Chao YC, Wu HM. The relationship between hot flashes and fatty acid binding protein 2 in postmenopausal women. PLoS ONE. 2022;17(10):e0276391. 10.1371/journal.pone.0276391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, et al. The clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022;33(10):2049–102. 10.1007/s00198-021-05900-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Stepan JJ, Hruskova H, Kverka M. Update on Menopausal Hormone Therapy for Fracture Prevention. Curr Osteoporos Rep. 2019;17(6):465–73. 10.1007/s11914-019-00549-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.