Next Article in Journal
The Role of Macrophages in the Pathogenesis of Atherosclerosis
Previous Article in Journal
Gene-Edited Human-Induced Pluripotent Stem Cell Lines to Elucidate DAND5 Function throughout Cardiac Differentiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cellular and Molecular Mechanisms Associating Obesity to Bone Loss

by
Yasmin Silva Forte
1,
Mariana Renovato-Martins
2 and
Christina Barja-Fidalgo
1,*
1
Laboratory of Cellular & Molecular Pharmacology, IBRAG, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
2
Laboratory of Inflammation & Metabolism, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil
*
Author to whom correspondence should be addressed.
Cells 2023, 12(4), 521; https://doi.org/10.3390/cells12040521
Submission received: 18 November 2022 / Revised: 1 February 2023 / Accepted: 3 February 2023 / Published: 5 February 2023
(This article belongs to the Section Tissues and Organs)

Abstract

:
Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.

Graphical Abstract

1. Introduction

Obesity is a disease characterized by abnormal or excessive fat accumulation that can harm health [1]. By 2030, it is estimated that over 1 billion people globally will become obese, with 20% in the female population and 15% in the male population [2]. Changes in society eating habits, following Western diet patterns, and reduced physical activity support its worldwide spread [3]. Obesity is associated with many diseases, such as diabetes, osteoarthritis, cardiovascular disorders, and some types of cancers, contributing to a further rise in the mortality rate [4,5,6]. It is estimated that 2.8 million people die annually from being overweight and obese [7].
The onset and development of the obesity condition depends on microenvironmental, metabolic, genetic, and behavioral factors [3]. The imbalance between increased food intake and decreased energy expenditure leads to alterations in adipose tissue distribution and secretion [8,9]. The hypertrophy of adipocytes, caused by excessive fat accumulation, enhances the expression of proinflammatory mediators, such as TNF-α, IL1β, IL6, resistin, and leptin in the adipose tissue. Infiltration of M1-like macrophages and hypoxic conditions due to insufficient blood vessel availability favors adipose tissue inflammation [10,11,12]. The release of adipose tissue-derived mediators in the bloodstream promotes a systemic low-grade inflammatory state that may reach other sites, such as the hypothalamus, skeletal muscle, pancreas, liver, and bone [13,14,15,16,17,18].
The obese adipose tissue also secretes increasing levels of extracellular vesicles (EVs) containing a variety of molecular cargos that may contribute to systemic alterations [19]. These EVs provide cellular communication, acting paracrine and endocrine, through the delivery of different proteins, transmembrane receptors, lipids, and nucleic acids [20,21]. We have recently demonstrated that the obese human adipose tissue releases more significant amounts of EVs than eutrophic adipose tissue [22]. Those EVs may regulate physiological and pathological processes, such as bone remodeling, cancer progression, embryonic development, tissue repair, and other biological phenomena [21,23,24]. The obese adipose tissue vesicles elicit a proinflammatory phenotype in monocytes and increase breast cancer malignancy [22,25]. However, little is known about obese adipose tissue EV content and their participation in bone remodeling. During obesity, non-coding RNAs (Nc-RNA) are released within extracellular vesicles or freely and contribute to systemic alterations. Nc-RNAs have been suggested as putative tools for diagnostics and therapeutics [26,27]. Nevertheless, the role of Nc-RNAs content from obese adipose tissue VEs on bone remodeling is still poorly investigated.
Bone is a dynamic calcified tissue involved in physical support, visceral protection, mineral storage, hematopoiesis, and endocrine activity [28,29,30]. Bone tissue renewal is a very balanced process where hormones, cytokines, and growth factors regulate bone formation and bone resorption [31,32]. Osteoblasts are bone-forming cells derived from mesenchymal stem cells (MSCs). They produce organic extracellular matrix elements and promote their further mineralization [33,34]. On counterbalance, osteoclasts are multinucleated cells derived from the monocyte/macrophage lineage, able to resorb bone by enzymatic digestion. Bone resorption occurs predominantly by cathepsin-K and MMP-9 actions in an acidic microenvironment [35,36]. A fine balance between osteoclast and osteoblast activities controls healthy bone remodeling [37]. Additionally, osteocytes are bone cells sensitive to mechanical loads that orchestrate regulatory mechanisms to enable bone turnover. The process involves the activation of signaling routes, such as WNT, and the release of mediators, such as bone morphogenetic proteins (BMP), nitric oxide (NO), prostaglandin E2 (PGE2), sclerostatin, FGF-23 and RANKL [38,39].
Due to the systemic inflammatory condition, obesity can disrupt the balance of bone remodeling, dysregulating bone homeostasis and allowing bone loss. Two main pathways drive this condition: (i) the release of proinflammatory mediators from obese adipose tissue that reduces osteoblast activity while increasing osteoclast differentiation and resorption; and (ii) the differentiation of MSCs towards osteogenic lineage is reduced during obesity, while adipogenic differentiation is enhanced [40,41,42]. Nevertheless, other systemic alterations may also have relevance in this process, including oxidative stress, gut and microbiota physiology, vesiculated or free Nc-RNA, and hormonal changes. This review gathered the main mechanisms involved in the crosstalk between obese adipose tissue and bones that lead to bone loss.

2. Materials and Methods

The following keywords, in combination or isolated forms, were searched on PubMed and Google Scholar platforms: “Obesity”; “TNF-α”; “IL-1β”, “IL-6”; “Leptin”; “Adiponectin”; “Lipocalin 2”; “gut microbiota”; “Lipopolysaccharide (LPS)”; “Reactive Oxygen Species (ROS)”; “osteocalcin”; “glucose metabolism”; “osteoblast”; “osteoclast”; “bone loss”; “NcRNA” and “MiRNA.” Reviews, repeated studies, and articles with divergent subjects were excluded. Experimental and cohort analyses published recently and suitable for our research were included. The figures were created using LibreOffice IMPRESS 4.7 program, and a few images from the copyright-free image websites PIXABAY and FLATICON were used for composition.

3. Results

3.1. Proinflammatory Cytokines in Bone Metabolism

Adipose tissue expansion increases pro-inflammatory cytokine expression and secretion by hypertrophic adipocytes and infiltration of M1-like macrophages and other immune cells (Figure 1). Cytokines, such as TNF-α, IL-1β, and IL-6 released by obese adipose tissue can interfere with bone cell homeostasis. Acting in isolation or in concert, they trigger intracellular signaling pathways that may lead to bone loss.
TNF-α is a well know enhancer of osteoclastogenesis [43]. In vitro experiments showed that stimulation of osteoclast precursors with TNF-α and RANKL increased the number of multinucleated TRAP-positive cells [44,45]. Additionally, in vivo treatment with TNF-α increased the osteoclast numbers in mice calvaria [46]. In this context, Shinohara and collaborators (2015) demonstrated that TNF-α-induced osteoclast formation depends on double-stranded RNA-dependent protein kinase [44]. Other studies showed that TNF-α increased c-kit+/c-fms+ osteoclast precursors in high-fat diet (HFD) fed obese mice and augmented RANK expression in bone marrow macrophages [47,48]. Increased RANKL expression in TNF-α stimulated osteocytes contributed to osteoclast differentiation in vitro and in vivo [43].
In parallel, TNF-α can also modify osteoblast activity: In MC3T3-E1, osteoblasts or MSCs induced osteogenic differentiation, TNF-α decreased the expression of osteogenic transcription factors RUNX2 and Osterix, reducing the mineralization and expression of bone markers, osteocalcin (OCN), alkaline phosphatase (ALP), and bone sialoprotein (BSP) [49,50,51,52,53]. These effects are mediated by the inflammatory transcription factors ATF3 [51] and Nf-κB [53]. TNF-α upregulated miR-150-3p in osteoblasts, decreasing β-catenin expression, a key transcription factor for osteogenesis [52]. This cytokine can affect osteoblast viability, promoting apoptosis [54] and increasing MMP-9 expression [55]. TNF-β, another TNF family member, also showed deleterious impacts on bones, reducing RUNX2, Type I collagen (Col1A), OCN, and integrin β1 expression and decreasing mineralization in MSCs cultured in the osteogenic medium [56].
In vivo studies have demonstrated TNF-α-related bone loss in obese mice [57,58]. A palmitic acid-enriched HFD enhanced TNF-α serum levels in obese mice, parallel with a decrease in bone turnover markers, P1NP e CTX1 [57]. Furthermore, mice fed with a HFD presented lower trabeculae numbers and thickness and reduced trabecular bone volume, compared to lean controls. Obese TNF-α knockout mice showed less bone loss, a reduced number of femoral osteoclasts, and enhanced RUNX2 expression in MSCs, compared to HFD wildtype [58].
Increased circulating levels of IL-1β are a hallmark of the chronic, low-grade inflammation associated with obesity and related diseases [59]. IL-1β is a known inducer of osteoclast migration and resorption [60,61,62]. Combined with RANKL, IL-1β significantly increased osteoclast formation in vitro, enhancing TRAP staining and the expression of osteoclast markers, cathepsin K, OSCAR, NFATC1, Cfos, and DC-STAMP [61,62,63,64,65]. Though IL-1β increases osteoclast formation, its effect depends on the precursor subsets. Comparing the early myeloid blasts and monocytes, IL-1β increased osteoclast formation and cell diameter, mostly in myeloid blasts [61]. IL-1β can also harm other bone cells, inducing apoptosis in the MLO-Y4 osteocytic cell line [66] and MC3T3-E1 osteoblasts [67]. Osteoblasts stimulated with IL-1β exhibited a lower migration capacity [68] and released more IL-6 [69]. Indirectly, IL-1β promoted an osteoclastic supportive phenotype in osteocytes, enhancing RANKL expression [67]. Apart from triggering specific mechanisms, IL-1β also shares some signaling mechanisms with TNF-α. Lee and collaborators (2017) have shown that treatment with IL-1β and TNF-α raised CCR7 expression in both precursors and differentiated osteoclasts, increasing their migratory activity toward CCL19 and CCL21 chemokines upregulated in rheumatoid arthritis [60]. Adseverin, an actin-binding protein that regulates cell differentiation and fusion, modulates IL-1β- and TNF-α-induced osteoclastogenesis [63]. Nevertheless, Hah and collaborators (2013) showed that both IL-1β and TNF-α increased ALP activity and mineralization of periosteal osteoblasts without modifying RUNX2 and OCN expression [70].
Although osteoblasts are characterized by their activity of extracellular matrix synthesis, IL-1β enhanced metalloprotease production by osteoblasts. Ozeki and collaborators (2014) demonstrated that this cytokine potentiated MMP13 expression through ADAM28 upregulation in osteoblast-like cells [67]. Yang and collaborators (2011) have shown that IL-1β amplified the expression of MMP-9 and MMP-13 in osteoblasts [71], further contributing to bone destruction in inflammatory diseases. IL-1β may also interfere with hormonal factors produced by bone tissue, such as FGF23, which orchestrates vitamin D and phosphate serum levels [72,73]. An imbalance in FGF23 regulation, mainly due to its exceeding action, leads to a pathological mineralization process that weakens bones [74]. Corroborating in vivo studies that described greater serum levels of FGF23 in IL-1β treated mice [73], the treatment of bone slices in vitro increased hormone secretion [72]. Additionally, He and collaborators (2020) demonstrated an association between IL-1β genetic variants and osteoporosis predisposition [75]. IL-1β variants rs1143627, rs16944, and rs1143623 are related to elevated susceptibility to osteoporosis, especially in women older than 60 or with a BMI greater than 24 kg/m2 [75]. In obese mice fed a 10% corn oil-based diet, Halade and colleagues (2011) observed that bone marrow adipose tissue secreted higher concentrations of IL-1β, TNF-A, and IL-6, which increased the expression of RANK-L in stromal cells favoring osteoclast formation [76].
IL-6 is a cytokine with pleiotropic actions secreted by several cell types, including adipocytes, and its plasma levels are significantly upregulated during obesity [77]. In bone tissue, such as IL-1β and TNF-α, IL-6 indirectly stimulated osteoclastogenesis, increasing RANKL expression in osteocytes and osteoblasts [78,79,80]. Using an in vitro model of bone loss, neutralizing IL-6 antibodies had protective effects against osteoporosis, enhancing bone mineral density, trabecular number, and thickness [81]. IL-6 inhibition decreased osteoblasts’ RANKL/OPG ratio and osteoclast differentiation in a microgravity model [81]. IL-6 also inhibited osteoblast activity by downregulating the expression of the osteogenic transcription factor RUNX2 [79,81,82]. IL-6 knockout osteoblasts presented higher ALP activity and RUNX2 expression than wild-type ones. Moreover, obese IL-6 knockout mice exhibited increased trabecular bone volume, number, and thickness than HFD wild-type mice [82].
Few publications have described a combined effect of proinflammatory cytokines on bone cells. Stimulation with IL-1β, TNF-α, and IL-6 together in bone chips enhanced IL- 1β, IL-6, IL-8, TNF- α, FGF23, SOST, and OPG expression by bone cells [83]. Yokota and collaborators (2013) showed that TNF-α and IL-6 supracalvarial injections in mice raised osteoclast formation, compared to isolated injections of both cytokines [84]. Together, these data suggest the pro-osteoclastic and anti-osteoblastic effects of proinflammatory cytokines and their importance in obesity-associated bone loss (Figure 2).

3.2. Adipokines in Bone Remodeling

Leptin and adiponectin are the two main characteristic adipokines produced by adipose tissue [85]. Leptin is also synthesized by the gastrointestinal tract, muscle, and brain [86] and regulates food intake, body weight, reproductive system, proinflammatory responses, and lipid metabolism [87]. Adiponectin is mostly synthesized by the adipocytes but also by myocytes and endothelial cells, acting to increase insulin sensibility, fatty acid oxidation, and anti-inflammatory, anti-diabetic, and anti-atherogenic effects [88,89]. Evidence has demonstrated that leptin and adiponectin have regulatory effects on bone metabolism [90,91].
Increased adiponectin and lower leptin secretion characterize homeostasis of healthy lean adipose tissue. In contrast, in obesity, the pathological adipose tissue expansion, with hypertrophy of adipocytes, unbalance that pattern, and the obese adipose tissue releases more leptin and less adiponectin [85,92]. These alterations contribute to systemic inflammation, insulin resistance, and skeletal disturbances during obesity [93,94].
The relevance of adiponectin signaling on bone tissue homeostasis has been evidenced in mice knockout models and cell culture systems. The deficiency of adiponectin in vivo resulted in bone loss, characterized by decreased bone volume/tissue volume ratio in the number of trabeculae, and the increase in trabecular separation [95], without alteration in bone mineral density [96]. Lack of adiponectin also compromised osteointegration of bone explants in deficient mice [97]. Abbott and collaborators (2015) described a reduction of 15% in the tibial fractional bone volume of mice with suppression of adiponectin in adipocytes, compared to controls, probably due to a higher differentiation or proliferation of bone marrow adipocytes in those animals [98]. Adiponectin seems to have a determinative impact on osteoclast formation and function. A decrease in adiponectin availability enhanced the number of osteoclasts in the femur of knockout mice [96,97]. The treatment of osteoclast precursors with recombinant adiponectin reduced multinucleated TRAP+ cell formation and expression of osteoclast markers, such as cathepsin K, OSCAR, and NFAT2 [97,98,99,100,101,102,103]. It Interfered in osteoclastogenesis signaling pathways, such as NF-κB, MAPK, and AKT [8,9]. Adiponectin reduced the viability of osteoclast progenitors, as shown by apoptosis and proliferation assays [97,101].
Unlike osteoclasts, adiponectin improved osteoblasts’ functions. Adiponectin treatment increased Osterix and BSP promoter activities in MC3T3-E1 murine osteoblasts [97] and enhanced extracellular matrix mineralization of primary osteoblasts and pre-osteoblasts [102,104]. Pacheco-Pantoja and collaborators (2014) demonstrated a reduced ALP activity in SAOS-2 osteoblasts [105]. Much evidence has demonstrated the role of adiponectin in osteoblast/adipocyte differentiation balance. In a model of ovariectomy-induced bone loss, the delivery of adiponectin by an adenovirus system raised mRNA levels of OCN, RUNX2, and ALP and reduced PPARγ and CEBP on bone tissue [106]. Adiporon, an agonist of adiponectin, favored osteogenic differentiation of bone marrow- and adipose tissue-derived MSCs, in vitro and in vivo, while decreasing adipogenic differentiation [102].
Adiponectin gene expression positively correlated with RUNX2 and negatively with RANKL/OPG ratio in the bones of healthy and osteoporotic patients [107]. In multiple myeloma patients, adiponectin was positively correlated with OCN, a bone formation marker, and negatively with the resorption marker CTX-1 [99]. Hence, adiponectin deficiency may be an essential mechanism of bone remodeling disbalance, including resorption stimulation and inhibition of bone formation in subjects with obesity.
In contrast with the consolidated data obtained with adiponectin studies, the effects of leptin on bone tissue are still conflicting. The contrasts occurred mainly between experimental and human models. Yue and collaborators (2016) showed that obese leptin receptor knockout mice presented an increased trabecular number and thickness, mineral apposition and formation rates, and decreased bone marrow adipocyte number, compared to the wild-type obese group [108]. Supporting the data showing a link between leptin and bone resorption, rats submitted for eight weeks to a HFD exhibited a lower trabecular number/thickness and bone mineral density (BMD), in parallel with serum leptin levels positively correlating with RANK levels [109]. In an innovative model, Carnovali and collaborators (2017) showed that adult male zebrafish fed a HFD presented increased ratio leptin/adiponectin serum levels, accompanied by a 12.6% reduction of the bone mineralized area, decrease in ALP activity, and increased TRAP activity and analog RANKL expression, compared to animals fed a standard diet [110]. In contrast, Feresin et al. (2014) did not find differences in bone parameters between Zucker rats fed a normal or HFD [111]. Philbrick and collaborators (2017) showed that administration of leptin in ob/ob mice increased bone formation rates and CTX-1 serum levels [112] and accelerated bone repair after the surgical fracture model [113].
Discordant information is also found in studies on the role of leptin in MSCs. While Tencerova and collaborators (2019) found that LepR+ bone marrow-MSCs from patients with obesity are associated with accelerated senescence and bone fragility [114], Li’s group showed that human MSCs differentiated toward osteogenic lineage in the presence of leptin had an increased ALP activity, compared with untreated cells [115]. Furthermore, analysis in a cohort of adolescents has demonstrated that bone mineral density (BMD) was positively correlated with serum leptin and fat mass [116]. Similarly, lumbar and hip BMD of Japanese postmenopausal women presented a positive correlation with leptin and a negative one with adiponectin [117]. Moreover, studies comparing children with normal weight and with obesity concluded that leptin levels are inversely related to tibial trabecular thickness [118].
Conflicting data may be explained by the distinct signaling pathways activated by leptin interfering differently in trabecular and cortical bone formation. Leptin regulates bone metabolism through direct anabolic signaling on bone cells and indirectly by the central nervous system. In the hypothalamus, leptin-mediated signaling regulates downstream effectors, such as neuropeptide-Y. In the presence of leptin, neuropeptide Y activity is inhibited, which causes an increase in cortical bone mass [119]. Wong and collaborators (2013) showed that knockout mice for leptin and neuropeptide Y have more significant gains in the cortical bone mass of long bones and vertebrae and a better mineral apposition rate than ob/ob mice [120].
Interestingly, in obese neuropeptide-Y-knockout mice, the cortical bone loss and fat accrual were suppressed, compared to wild types [121]. This information is relevant because, in subjects with obesity with leptin receptor resistance, the lack of signaling in bone cells and the reduction or inhibition of neuropeptide Y can contribute to deleterious effects on bones. However, the exact consequence of leptin signaling on bone tissue in obesity is still unclear.
In addition to leptin and adiponectin, ghrelin and glucagon-like peptide 1 (GLP-1), are altered during obesity and also influence bone metabolism [122], and may have a role in bone remodeling. Ghrelin is produced by the stomach, acting on appetite regulation and energy metabolism, improves the cardiovascular function, and can act in bone formation [123]. Erener et al. (2021) showed that ghrelin administration in mice enhanced the healing process of bone fractures [123]. Notably, in patients with osteopenia, the systemic ghrelin concentration are inversely related to BMI values, and the osteoblasts isolated from those patients were resistant to ghrelin-mediated signaling [124].
The therapeutic use of GLP-1 receptor agonists has been well described to improve for the metabolic parameters of patients with obesity and insulin resistance [125]. Additionally, activation of GLP-1 receptor on MSCs induced osteogenic differentiation via PKA/β-catenin signaling activation [126], and improved bone composition in obese animals [127]. Together, the data point to ghrelin and GLP-1 as worthy candidates for pharmacological intervention for controlling obesity-associated bone loss.

3.3. Muscle-Fat-Bone Crosstalk during Obesity

Bones and skeletal muscles are associated tissues that promote locomotion, and the mechanical forces driven by muscle contraction are vital factors that allow bone formation. However, the muscle-bone crosstalk is not limited to structural functions. Myokines produced and released in response to muscular contractions have osteoanabolic effects and are essential for maintaining bone metabolism [128]. Chronic low-grade inflammation in patients with obesity modifies the crosstalk between muscle-fat-bone [129]. Fatty acids released by intramuscular adipose tissue induced lipotoxic effects on osteoblasts, osteocytes, and myocytes [130]. Disturbances in muscle metabolism caused by overweight/obesity may impact bone tissue, contributing to osteosarcopenic obesity syndrome.
The sedentary profile of people with obesity reduces the anti-inflammatory activity performed by myokines, such as irisin. This exercise-triggered myokine promotes white adipose tissue browning, energy expenditure and stimulates bone formation [131]. Knockout mice for the irisin precursor FNDC5 (fibronectin-type III domain-containing 5) showed bone loss mediated by increased resorption activity and reduced bone formation [132,133]. Irisin stimulates osteoblast differentiation through αV integrin activating-BMP/SMAD pathway [132], leading to activation of RUNX1/2 via focal adhesion kinase [133]. The administration of irisin in high-fat diet-induced obese mice increased insulin, ghrelin, and BMP4 circulating concentrations [134]. Obese rodents subjected to swimming showed improvement in bone loss, simultaneously with increased systemic irisin production and release [135]. Addressing the crosstalk among bone, muscle, and adipose tissue may be a successful pharmacological strategy for treating bone and muscle loss in obesity.

3.4. Gut and Microbiota Alterations Influencing Bone Metabolism

The crosstalk between obesity and intestinal disorders still raises doubts about the cause and effect mechanisms since weight gain and intestinal damage are complementary events. Studies using germ-free mice proved the importance of gut microbiota regulation on fat accrual and obesity development, where a lack of microorganisms reduced 42% of total body fat [136]. Additionally, microbiota transplantation from obese to germ-free mice increased body fat, compared to lean microbiota transplantation [137]. Concurrently, it is well known that environmental factors, such as the Western diet compromise gut microbiota quality, favoring weight gain [138]. In individuals with obesity, proinflammatory cytokines, such as TNF-α and IFN-γ produced by adipose tissue interfere with tight junctions of the intestinal barrier, promoting increased permeability [139]. As a result, components of gram-negative bacteria cell wall, such as the endotoxin lipopolysaccharide (LPS), can bypass the loosened intestinal cell junctions and enters the bloodstream. Enhanced LPS serum levels contribute to the exacerbation of inflammation, insulin resistance, adipose tissue pathological expansion, and other systemic damage [140]. Therefore, obesity interferes with bone homeostasis through gut modifications in two principal ways: The first is the change in the intestinal microbiota profile, and the second is the elevated serum endotoxin levels (Figure 3).
Luo and collaborators (2015) demonstrated that fecal microbiota transplantation (FMT) from HFD mice to normal diet mice decreased the osteoblast surface and RUNX2 gene expression [141]. In a periodontitis model, FMT from obese mice also increased the periodontal tissue inflammation and bone destruction [142].
Among several types of bacteria in the gut microbiota, two main phyla stand out, Bacteroidetes and Firmicutes. In healthy individuals, there is usually a favoring in Bacteroidetes growth. However, subjects with obesity tend to have a more significant presence of Firmicutes [143]. Changes in Firmicutes/Bacteroidetes ratio have been associated with inflammatory conditions and systemic effects [144]. Mice fed a high-fat diet presented an enhancement of the Firmicutes/Bacteroidetes ratio, which correlated negatively with bone volume [145]. In a complementary way, another study comparing sham and ovariectomized mice showed that Bacteroidetes positively correlate with bone volume/tissue volume, bone mineral density, and trabecular number. At the same time, Firmicutes are negatively related to the same bone microstructure parameters [146]. Another phylum, Proteobacteria, also appears to be associated with obesity-bone loss. In a Mexican population study, children with obesity presented an abundance of Proteobacteria phylum, compared to normal-weight children in stool samples [147]. At the same time, more significant amounts of Proteobacteria were found in fecal samples of low BMD women, compared to average BMD women [148].
Along with Bacteroidetes, obesity reduces other beneficial bacteria genera, such as Akkermansia, Bifidobacterium, and Lactobacillus [149], compromising healthy bone remodeling. The beneficial bone effects of Akkermansia were shown to be mediated by EVs release, which enhanced osteoblast differentiation and mineralization while decreasing osteoclasts formation [150]. Akkermansia muciniphila administration reduced local inflammation and bone destruction in a model of infection-induced bone loss [151]. Moreover, patients with osteopenia had a lower frequency of Akkermansia bacteria in fecal samples than healthy patients, although the comparison did not establish a statistical difference [152].
Bifidobacterium pseudocatenulatum administration in HFD-induced obese mice improved the trabecular parameters, compared to non-treated obese mice. It increased the genic expression of Wnt1, a ligand that induces osteogenesis through the β-catenin signaling pathway. Serum dosages also showed that Bifidobacterium enhanced the bone formation marker OCN and decreased the bone resorption marker CTX-1 in HFD mice [153]. Corroborating this data, administration in obese mice of VSL#3, a commercial probiotic formulated predominantly by Bifidobacterium and Lactobacillus species, increased the trabecular bone volume and bone mechanical strength, compared to the obese vehicle group. Bone formation was due to the inhibitory effect upon osteoclast differentiation and the favoring of osteoblast activity through histone methylation [154]. The treatment of obese insulin-resistant mice with Lactobacillus paracasei, alone or conjugated with the prebiotic Xylooligosacharide (XOS), reduced systemic inflammation [155,156] and the eroded bone surfaces, reestablishing bone mineral apposition and formation, harmed by obesity [156]. It was reported that the probiotic Lactobacillus rhamnosus GG increases bone mass by modifying immune responses by enhancing serum butyrate concentrations. Butyrate raises the frequency of T-regulatory lymphocytes in the gut and bone marrow which, in return, stimulates TCD8+ Lymphocytes to release wnt10b, a positive regulator of bone formation [157]. Obesity promotes the reduction of butyrate-producing bacteria in the gut [158], which may be an additional interference with bone metabolism.
Due to obesity-induced dysbiosis, prebiotics have become an alternative therapy to revert the prejudicial impacts of imbalanced microbiota on different systems. In addition to XOS [155,156], other prebiotics, such as Fructooligosaccharide and Galactooligosaccharide reduced bone loss in HFD mice, increasing osteoblast differentiation and decreasing osteoclast and adipocyte formation. The prebiotics also ameliorates gut dysbiosis by reducing the Firmicutes/Bacteroidetes ratio, normalizing intestinal permeability [159]. Combined with exercise, fiber prebiotics prevented knee joint damage, insulin resistance, and endotoxemia in high-fat and high-sucrose diet (HFS) mice [160].
Changes in gut permeability observed during obesity amplify the availability of LPS—a potent inducer of bone degradation [155,160]. In animal models of bone loss, systemic (intraperitoneal) or local (in calvaria) administration of LPS promoted reduction in BMD, trabecular thickness, and bone volume while increasing the number of TRAP+ cells on the bone surface and CTX-1 serum concentration [161,162,163,164]. In female Zucker fat rats, LPS administration also decreased bone volume [164]. LPS stimulation of MC3T3-E1 osteoblasts reduced cell viability with an increased genic expression of caspase 3 and Bax [165], decreasing the ALP activity and expression of osteogenic markers [165,166,167]. Moreover, LPS treatment modifies osteoblasts to better support osteoclastogenesis. Primary osteoblasts from LPS-treated mice had enhanced expression of RANKL, compared to mice that received saline injections [163]. Moreover, LPS increased EphA2 expression in both osteoblasts and osteoclasts in vitro [166]. Signaling mediated by EphA2 and its ligand EphrinA2 promoted osteoclast formation and inhibited osteoblastogenesis [168]. Together with RANKL, LPS potentiated multinucleated TRAP+ cell differentiation in vitro and raised the expression of cathepsin K, MMP-9, and NFATc1 [162,166,167]. Xing and collaborators (2011) also showed that LPS promotes pre-osteoclasts migration and differentiation through CXCR4 expression via TLR4 [169]. This whole dataset suggests therapeutic strategies that target the resolution of obesity-induced gut dysbiosis and compromised intestinal barrier integrity as relevant ways to combat bone loss.

3.5. Osteocalcin, FGF23, and Lipocalin-2 Regulations upon Glucose Metabolism

Beyond their mechanical properties associated with physical support and visceral protection, the bone tissue has other functional activities, including mineral storage, hematopoiesis support, and hormone release [28]. The endocrine function of bones was recently discovered, and the three main osteokines produced are FGF23, osteocalcin (OCN), and lipocalin-2 [170]. FGF23 is produced by osteoblasts and osteocytes and regulates phosphate resorption by renal proximal tubules and calcitriol production [171,172]. OCN, a bone formation marker, stimulates insulin sensitivity and production and increases energy expenditure [173].
Similarly, lipocalin-2 regulates insulin signaling, improves glucose tolerance, and inhibits appetite [174]. Therefore, bone tissue plays a crucial role in maintaining a healthy energy metabolism. Disturbances in bone metabolism promoted by obesity interfere with glucose metabolism and weight gain, worsening obesity conditions and aggravating bone loss (Figure 4).
OCN is produced by mature osteoblasts and assumes different activities depending on post-translational modifications. γ-carboxylated osteocalcin (GlaOC), due to carboxylation, binds to hydroxyapatite and is part of the bone extracellular matrix. Uncarboxilated osteocalcin (UnOC), released after osteoclast’s bone resorption, works as a hormone and promotes glucose tolerance [173]. Studies comparing individuals with normal weight and with obesity showed that obesity reduces UnOC serum concentration, as seen in works that compared premenopausal women, children, and men with obesity with their respective lean controls [175,176,177,178]. Additionally, Vigevano and collaborators (2021) demonstrated that, compared to healthy men with obesity, type 2 diabetes-bearing men with obesity had a lower UnOC serum concentration, increased trabecular separation at the tibia and radius, and decreased tibial failure load and stiffness [179]. In HFD-induced obese mice, administration of UnOC reduces weight and fat pad gain [180,181], insulin resistance, and the expression of inflammatory genes [181]. Moreover, it improves glucose tolerance [180,181] and increases glucose transporter type 4 (Glut4) expression in white adipose tissue of treated animals [181]. UnOC injections also prevented obesity and type 2 diabetes in HFD mice and increased the number of mitochondria in skeletal muscle [182].
Unlike OCN, FGF23 serum concentrations have been positively correlated with the BMI [183,184]. In this context, studies including pre- and post-menopause women and adult men showed that circulating FGF23 was significantly higher in individuals with obesity, compared to lean individuals [185,186]. High FGF23 availability is associated with declined metabolic function and cardiovascular disorders [183,185]. Additionally, changes in this osteokine compromise bone metabolism. Rupp and colleagues (2019) demonstrated that high serum levels of FGF23 correlated with trabecular bone loss in osteoporotic patients [74]. Moreover, in an osteoporosis mouse model, FGF23 contributed to bone loss by activating the JAK/STAT pathway [187]. Although FGF23 is widely associated with phosphate and vitamin D metabolisms, some studies have implicated this hormone in insulin resistance [183,188].
Indeed, diabetes is an additional factor that promotes osteoporosis [189] and, combined with obesity, amplifies bone loss. Deleterious effects on the bones of diabetic subjects seem to occur mainly due to the inhibition of osteoblast activity. Under high glucose conditions, in vitro, osteoblasts present reduced expression of ALP, OCN, and RUNX2, while proinflammatory cytokines IL-1β and IL-6 were increased, which is an indirect way to stimulate osteoclasts formation [190]. In diabetic mice and in vitro models, high glucose concentrations decreased osteoblast proliferation and differentiation through pyroptosis [191]. In HFD-induced obese mice, metformin administration reversed the reduction of osteoblasts in long bones [192]. Subjects with obesity may present high serum concentrations of advanced glycation end-products (AGEs) in urine samples. Osteoblasts treated with AGEs presented lower OCN and Col1A expression [178]. Despite its effect on osteoblasts, the mechanisms explaining how glucose impacts osteoclasts are still not understood. Contradictory data shows both increase [193] and inhibition [194] of osteoclast differentiation and resorption activity face high glucose concentrations in vitro.
Obesity also influences serum lipocalin-2. Despite its action in improving insulin sensitivity and appetite inhibition [174,195], patients with obesity present greater serum levels of this hormone than lean individuals [196,197,198]. This intriguing data is justified as a compensatory mechanism trying to counter the metabolic disturbance promoted by obesity [195]. Hence, increased lipocalin-2 production by bones could be a protective response to diabetes-associated bone loss. This hypothesis is supported by studies revealing that lipocalin-2 negatively regulates osteoclast formation [199]. However, many studies describe the harmful effects of excessive lipocalin-2 on bone remodeling. Some works emphasize the catabolic and inflammatory responses triggered by lipocalin-2, which impact systemically [200]. In this way, an increase in lipocalin-2 was associated with an 80 to 81% increased risk of osteopenia and hip fracture [198].
Additionally, overexpression of lipocalin-2 in bones reduced the cortical bone layer and trabecular number and increased RANKL and IL-6 expression. An increased number of osteoclasts was also identified in vivo and in vitro upon lipocalin-2 stimulation, compared to controls [201]. Osteoblasts transfected with lipocalin-2 presented lower expression of RUNX2, Osterix, and ALP [202]. Therefore, it is necessary to elucidate the role of lipocalin-2 in obesity and its systemic effects on bones to establish robust conclusions. Moreover, insulin resistance, mediated partly by UnOC and FGF23 (bone hormones) alterations in subjects with obesity, contributes to the osteoporosis scenario in obesity.

3.6. Oxidative Stress and Bone Remodeling

Oxidative stress is an imbalance between oxidizing agents, reactive radical or non-radical species, and antioxidant agents. In pathological situations, such as obesity, and cardiovascular and neurological diseases, reactive species production is predominant while antioxidant defense mechanisms are insufficient or reduced. Therefore, the excess of unstable molecules causes structural damage to proteins, DNA, and lipids, compromising cell integrity and functionality and leading to cell death [203,204]. Obesity induces oxidative stress in different ways, including hyperglycemia, increased plasma free fatty acids (FFAs) and endothelial dysfunction, deficiencies in minerals and vitamins, and diet composition [205]. Mitochondrial dysfunction occurs in adipocytes and immune cells within obese adipose tissue and promotes increased production of reactive oxygen species (ROS) and lipid peroxidation products [204,206]. Oxidative stress is observed in rodent models of obesity, with increased concentrations of plasmatic ROS and thiobarbituric acid reactive substances (TBARS) [207]. In subjects with obesity, subcutaneous adipose tissue samples present higher ROS levels and increased mitochondrial damage than lean ones [208].
Indeed, the release of oxidative stress mediators promotes considerable damage to bone tissue, enabling the development of osteoporosis [209]. In MSCs, enhanced levels of intracellular ROS released by NOX4 favor adipogenesis [210]. Concurrently, in a coculture system of 3T3-L1 differentiated adipocytes and MC3T3-E1 osteoblasts, adipocytes release FFAs, which promote oxidative stress in osteoblasts. In this context, osteoblasts present reduced mineralization activity, lower RUNX2, Col1A, and OCN expression, and increased apoptosis [211]. These data demonstrate that redox imbalance favors the expansion of adipose tissue while impacting bone formation.
NOX2 (GP91PHOX) is the catalytic subunit of NADPH oxidase that is highly expressed in immune cells. In HFD-induced obese mice, increased ROS production is observed in the bone marrow and white adipose tissue. The majority of ROS is produced by NOX2, as demonstrated in NOX2-deficient obese mice [212]. In addition to its role in the inflammatory status of adipose tissue during obesity, NOX2 regulates bone metabolism. Compared with obese wild-type mice, obese NOX2-knockout mice presented higher BMD and lower pro-osteoclastic factors, RANKL and cathepsin K. The bone marrow precursors derived from NOX2-knockout mice showed a reduced capacity to differentiate into osteoclasts in vitro, compared to wild-type cells [213]. Corroborating this data, Kang and Kim (2016) showed the role of NOX2 in promoting osteoclast differentiation through NFATC1 upregulation [214]. The data indicate that signaling mechanisms involving NOX2 activity mediate bone resorption during obesity.
Deficiency in antioxidant responses also contributes to oxidative stress and, consequently, unbalancing bone remodeling during obesity, which decreases glutathione peroxidases (GPX) [207,215,216]. HFD-induced obesity lowers the GPX3 plasma concentration in mice [207] and GPX1 expression in the liver [215] and reduces serum glutathione [216]. Hu and collaborators (2021) described that GPX7 favored osteogenic differentiation of MSCs and inhibited adipogenic commitment [217]. Obese GPX7 knockout mice under HFD presented increased inguinal, gonadal, and mesenteric fat accrual and higher plasmatic FFA concentration than wild-type obese mice [218]. An in silico analysis predicted that GPX7 expression is downregulated by miR 335-5p, which is upregulated in white adipose tissue of obese mice [219]. Thus, GPX7 may be a relevant target for oxidative stress-associated bone loss. However, GPX activities in obesity and their relationship with bone tissue regulation must be better elucidated.
Antioxidants may be a potential therapeutic approach to reduce the negative impacts of oxidative stress promoted by obesity on bones. The administration of N-acetylcysteine (NAC), a glutathione precursor, in HFD mice contributed to increased bone mass, compared to non-treated obese animals. This effect was explained due to the raised reduced glutathione disposal and decreased osteoclast formation, both in vivo and in vitro [216]. Beneficial effects were also observed in HFD mice fed with folic acid (FA), which reduced osteoporosis, malondialdehyde (MDA), and FFAs levels in bone homogenates while increasing Nrf2 expression [220]. Nrf2 upregulation in osteoblasts reduced IL-6 secretion, decreasing osteoclast differentiation in vitro [221]. In addition, green tea polyphenol supplementation in obese mice stimulated both BMD/bone strength and GTX1 expression in the liver and diminished fat mass and proinflammatory mediators [215]. Sodium butyrate, an antioxidant, ameliorated the metabolic profile of obese rats, enhancing mitochondrial antioxidant enzymes and redox homeostasis and improving bone condition [222]. Together, the data reinforce that oxidative stress promoted by obesity is a critical mechanism in bone loss, becoming a target of pharmacological strategies to alleviate osteoporosis.

3.7. Free and Vesiculated Noncoding RNAs

In addition to the protein-coding function that RNAs can assume, non-coding RNAs (Nc-RNA), often underestimated, have recently gained notoriety for their transcriptional and post-translational regulatory capacity. Nc-RNA is a large group of RNA sequences divided into two groups: long non-coding RNA (Lnc-RNA) and short non-coding RNA (Snc-RNA). Snc-RNA is formed by MicroRNA (MiRNA), small interfering RNA (siRNA), small nucleolar RNA (snoRNA), ribosomal RNA (rRNA), transfer RNA (tRNA), and piwi-interacting RNA (piRNA). Inc-RNA group comprises competing endogenous RNA (CeRNA), circular RNA (CircRNA), long intergenic non-coding RNA (lincRNA), antisense RNA, and pseudogenes [223,224].
The MiRNAs are one of the main subclasses of Snc-RNAs and control the expression of specific proteins through pairing with messenger RNAs (mRNA). Once paired with a complementary mRNA, MiRNA can induce mRNA cleavage or repress its translation on ribosomes, which blocks some protein synthesis [27]. CeRNAs, a subclass of Lnc-RNA, directly control the activity of MiRNAs and act like a “sponge,” competitively sequestering MiRNAs and inhibiting their interaction with mRNAs, reestablishing the expression of silenced genes [225].
In addition to their physiological functions, Nc-RNAs also contribute to the development of obesity, cancer, cardiovascular, autoimmune, and bone diseases. These Nc-RNAs released in their free form or carried by extracellular vesicles (EVs), are considered valuable for diagnostic strategies [226,227,228,229,230]. Herein, we have focused on five MiRNAs and one CeRNA that are described to be up-or downregulated in obesity and able to interfere in bone remodeling, becoming potential ways to induce osteoporosis. All selected data are comprised in Table 1.
MiR-34a-5p is upregulated in the plasma of subjects with obesity and late-state osteoarthritis, compared with healthy controls; furthermore, it is also upregulated in the plasma and knee joint of HFD mice [231]. It is known that HFD mice released increased amounts of derived exosomes enriched in MiR-34a, contributing to adipose tissue inflammation and inhibiting the M2 polarization of macrophages [232]. Yuan and collaborators (2021) demonstrated that MiR-34a-5p inhibits MSCs’ osteogenic differentiation [233]. A study analyzing old Brazilian adults demonstrated that high circulating MiR-34a-5p was associated with lower BMD in the lumbosacral region [234]. MiR-155 was implicated in osteogenesis inhibition through the regulation of SMAD5 [235], and SOCS1 [236] expression was detected in exosomes derived from obese adipose tissue and promoted insulin resistance [237]. Microvesicles derived from HFD mice’s adipocytes also contain more copies of MiR-155 [238]. In contrast, MiR-503, described as an enhancer of osteoblast differentiation [239] and inhibitor of adipogenesis [240], is downregulated in the plasma of subjects with obesity and is associated with a poor metabolic state [241]. An exciting work from Li and collaborators (2019) described MiR-149-3p as a regulator of MSC fate, inhibiting adipogenesis and favoring osteogenesis; its overexpression decreased obesity-associated gene (FTO) expression [242]. However, it is still unclear whether MiR-149-3p is overexpressed/suppressed or unchanged in patients with obesity.
Regarding the impact on osteoclast formation, two MiRNAs upregulated during obesity stand out, MiR-142-5p and MiR-155 [237,238,243]. MiR-142-5p targets PTEN-mediated signaling and increases bone marrow macrophage differentiation toward osteoclasts [244]. This MiRNA also reduces MSCs migration through VCAM1, contributing to osteoporosis [245]. MiRNA MiR-155, besides its effect on osteoblast-adipocyte regulation, can enhance osteoclastogenesis, targeting leptin receptor LepR expression [246]. In contrast, Zhang and colleagues (2012) showed an inhibitory effect of MiR-155 on osteoclast formation through modulation of IFN-β expression [247].
Table 1. Obesity-associated non-coding RNAs and their impacts on bone remodeling. Obese adipose tissue releases non-coding RNAs that promote enhanced formation of osteoclasts and adipocytes while decreasing bone formation and osteoblast differentiation. These non-coding RNAs could be released freely or inside extracellular vesicles, derived mainly by adipocytes and macrophages from obese adipose tissue.
Table 1. Obesity-associated non-coding RNAs and their impacts on bone remodeling. Obese adipose tissue releases non-coding RNAs that promote enhanced formation of osteoclasts and adipocytes while decreasing bone formation and osteoblast differentiation. These non-coding RNAs could be released freely or inside extracellular vesicles, derived mainly by adipocytes and macrophages from obese adipose tissue.
Noncoding RNATargetEffect on Bone CellsObesity StatusFree or Vesiculated
MiR-34a-5pWNT1 [233]Inhibition of Osteogenesis [233]
Decreased Bone Mineral Density [234]
Upregulated [231,232]Free [231]
Obese Adipocyte-derived Exosomes [232]
MiR-142-5pVCAM-1 [245]
PTEN [244]
Inhibits Bone Marrow MSCs Migration Leading to Osteoporosis [245]
Favors Osteoclast Differentiation [244]
Upregulated [243]Free
MiR-155SMAD5 [235]
SOCS1 [236]
LepR [246]
IFN-β [247]
Inhibit Osteogenesis [235,236]
Favors Osteoclast Differentiation [246]
Inhibits Osteoclast Differentiation [247]
Upregulated [237,238]Obese Adipose Tissue Macrophages-Derived Exosomes [237]
Obese Adipocyte-Derived Microvesicles [238]
MiR-503BMPR1a [240]
SMURF1 [239]
Inhibit Adipogenesis [240]
Favors Osteogenesis [239]
Downregulated [241]Free
Lnc-H19Mir-467/
HOX10 [248]
Favors Osteogenesis [248]
Inhibits Adipogenesis [248]
Downregulated [248]Mesenchymal Stem Cell-Derived Exosomes
MiR-149-3pObesity-Associated Gene FTO [242]Favors Osteogenesis [242]
Inhibit Adipogenesis [242]
Unknown
Overexpression of FTO Gene is Associated with Obesity Development [249]
Unknown
A CeRNA, Inc-H19, contributed to obesity-associated bone loss, acting as a competitive inhibitor binding to complementary MiRNAs, and blocking its actions. It was shown that HFD mice presented lower amounts of MSC-derived exosomes, with less Inc-H19 content than normal diet mice. Exosomes containing Lnc-H19 promoted osteogenesis and repressed adipogenesis in vitro. This action was mediated through MiR-467 inhibition, which modulates HOX10 expression [248].
In the set, the data highlight that during obesity, four MiRNAs and one Lnc-RNA, released by adipose tissue, contribute as additional mechanisms of bone loss by repressing osteogenesis and favoring adipogenesis and osteoclast formation. Additional studies should be conducted to identify new classes of Nc-RNAs in subjects with obesity that can interfere with bone metabolism and may contribute to developing new bone loss biomarkers and therapies for obesity osteoporotic state.

4. Conclusions

Obesity is an inflammatory disease that may promote loss of quality of life. The release of several proinflammatory factors by the adipose tissue induces the onset of other pathological conditions, such as diabetes, atherosclerosis, osteoarthritis, and cancers. In bones, obesity unbalances osteoblast and osteoclast activities leading to bone loss. Among the mechanisms contributing to obesity-associated osteoporosis, the following stand out: proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and adipokines (leptin and adiponectin); changes in gut microbiota and intestinal permeability; insulin resistance; oxidative stress; and free or EV-carried non-coding RNAs. The assessment of all of these mechanisms is relevant for understanding the harmful effects of obesity and developing new anti-obesity and anti-osteoporosis therapeutic strategies.

Author Contributions

The conception and design were performed by Y.S.F., M.R.-M., and C.B.-F.; financial support was obtained by C.B.-F. and Y.S.F. who also conducted the collection and/or assembly of data; Y.S.F., M.R.-M. and C.B.-F. performed the data analysis and interpretation; prepared the manuscript; and Y.S.F., C.B-F. and M.R.-M. supervised the study. The final approval for the manuscript: C.B.-F. and M.R.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ: C.B-F. E-26/200.898/2021; E-26/210.110/2020); Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq: C.B.-F. # 305737/2021-0); Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES-PROEX).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. The World Health Organization (WHO). Heath Topics Obesity. Available online: https://www.who.int/health-topics/obesity#tab=tab_1 (accessed on 12 February 2022).
  2. World Obesity Atlas 2022. Available online: https://www.worldobesity.org/resources/resource-library/world-obesity-atlas-2022 (accessed on 12 February 2022).
  3. Rogero, M.M.; Calder, P.C. Obesity, Inflammation, Toll-Like Receptor 4 and Fatty Acids. Nutrients 2018, 10, 432. [Google Scholar] [CrossRef] [PubMed]
  4. Piché, M.E.; Tchernof, A.; Després, J.P. Obesity Phenotypes, Diabetes, and Cardiovascular Diseases. Circ. Res. 2020, 126, 1477–1500. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, T.; He, C. Pro-inflammatory cytokines: The link between obesity and osteoarthritis. Cytokine Growth Factor Rev. 2018, 44, 38–50. [Google Scholar] [CrossRef]
  6. Avgerinos, K.I.; Spyrou, N.; Mantzoros, S.C.; Dalamaga, M. Obesity and cancer risk: Emerging biological mechanisms and perspective. Metab. Clin. Exp. 2019, 92, 121–135. [Google Scholar] [CrossRef] [PubMed]
  7. The World Health Organization (WHO) Obesity 2021. Available online: https://www.who.int/news-room/facts-in-pictures/detail/6-facts-on-obesity (accessed on 12 February 2022).
  8. Vecchié, A.; Dallegri, F.; Carbone, F.; Bonaventura, A.; Liberale, L.; Portincasa, P.; Frühbeck, G.; Montecucco, F. Obesity phenotypes and their paradoxical association with cardiovascular diseases. Eur. J. Intern. Med. 2017, 48, 6–17. [Google Scholar] [CrossRef] [PubMed]
  9. Unamuno, X.; Gómez-Ambrosi, J.; Rodríguez, A.; Becerril, S.; Frühbeck, G.; Catalán, V. Adipokine dysregulation and adipose tissue inflammation in human obesity. Eur. J. Clin. Investig. 2018, 48, e12997. [Google Scholar] [CrossRef]
  10. Sun, K.; Kusminski, C.M.; Scherer, E.P. Adipose tissue remodeling and obesity. J. Clin. Investig. 2011, 121, 2094–2101. [Google Scholar] [CrossRef]
  11. Deng, T.; Lyon, C.J.; Bergin, S.; Caligiuri, M.A.; 4 Hsueh, W.A. Obesity, Inflammation, and Cancer. Annu. Rev. Pathol. Mech. Dis. 2016, 11, 421–449. [Google Scholar] [CrossRef]
  12. Trayhurn, P. Hypoxia and Adipose Tissue Function and Dysfunction in Obesity. Physiol. Rev. 2013, 93, 1–21. [Google Scholar] [CrossRef]
  13. Stolarczyk, E. Adipose tissue inflammation in obesity: A metabolic or immune response? Curr. Opin. Pharmacol. 2017, 37, 35–40. [Google Scholar] [CrossRef]
  14. Thaler, J.P.; Yi, C.; Schur, E.A.; Guyenet, S.J.; Hwang, B.H.; Dietrich, M.O.; Zhao, X.; Sarruf, D.A.; Izgur, V.; Maravilla, K.R.; et al. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Investig. 2012, 122, 153–162. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, H.; Ballantyne, C.M. Skeletal muscle inflammation and insulin resistance in obesity. J. Clin. Investig. 2017, 127, 43–54. [Google Scholar] [CrossRef] [PubMed]
  16. Stern, J.H.; Smith, G.I.; Chen, S.; Unger, R.H.; Klein, S.; Scherer, P.E. Obesity dysregulates fasting-induced changes in glucagon secretion. J. Endocrinol. 2019, 243, 149–160. [Google Scholar] [CrossRef]
  17. Van der Heijden, R.A.; Sheedfar, F.; Morrison, M.C.; Hommelberg, P.P.H.; Kor, D.; Kloosterhuis, N.J.; Gruben, N.; Youssef, S.A.; de Bruin, A.; Hofker, M.H.; et al. High-fat diet induced obesity primes inflammation in adipose tissue prior to liver in C57BL/6j mice. Aging 2015, 7, 256–268. [Google Scholar] [CrossRef] [PubMed]
  18. López-Gómez, J.J.; Castrillón, J.L.P.; de Luis Román, D.A. Influencia de la obesidad sobre el metabolismo óseo. Endocrinol. Y Nutr. 2016, 63, 551–559. [Google Scholar] [CrossRef]
  19. Kim, A.; Shah, A.S.; Nakamura, T. Extracellular Vesicles: A Potential Novel Regulator of Obesity and Its Associated Complications. Children 2018, 5, 152. [Google Scholar] [CrossRef]
  20. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef]
  21. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  22. Renovato-Martins, M.; Matheus, M.E.; de Andrade, I.R.; Moraes, J.A.; da Silva, S.V.; dos Reis, M.C.; de Souza, A.A.P.; da Silva, C.C.; Bouskela, E.; Barja-Fidalgo, C. Microparticles derived from obese adipose tissue elicit a pro-inflammatory phenotype of CD16 +, CCR5 + and TLR8 + monocytes. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2017, 1863, 139–151. [Google Scholar] [CrossRef]
  23. Liu, M.; Sun, Y.; Zhang, Q. Emerging Role of Extracellular Vesicles in Bone Remodeling. J. Dent. Res. 2018, 97, 859–868. [Google Scholar] [CrossRef]
  24. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed]
  25. Ramos-Andrade, I.; Moraes, J.; Brandão-Costa, R.M.; Da Silva, S.V.; De Souza, A.; Da Silva, C.; Renovato-Martins, M.; Barja-Fidalgo, C. Obese adipose tissue extracellular vesicles raise breast cancer cell malignancy. Endocr.-Relat. Cancer 2020, 27, 571–582. [Google Scholar] [CrossRef]
  26. Ji, C.; Guo, X. The clinical potential of circulating microRNAs in obesity. Nat. Rev. Endocrinol. 2019, 15, 731–743. [Google Scholar] [CrossRef]
  27. de Sousa, M.C.; Gjorgjieva, M.; Dolicka, D.; Sobolewski, C.; Foti, M. Deciphering miRNAs’ Action through miRNA Editing. Int. J. Mol. Sci. 2019, 20, 6249. [Google Scholar] [CrossRef] [PubMed]
  28. Florencio-Silva, R.; da Silva Sasso, G.R.; Sasso-Cerri, E.; Simões, M.J.; Cerri, P.S. Biology of Bone Tissue: Structure, Function, and Factors That Influence Bone Cells. BioMed. Res. Int. 2015, 2015, 421746. [Google Scholar] [CrossRef]
  29. Czekanska, E.; Stoddart, M.; Richards, R.; Hayes, J. In search of an osteoblast cell model for in vitro research. Eur. Cells Mater. 2012, 24, 1–17. [Google Scholar] [CrossRef] [PubMed]
  30. Guntur, A.R.; Rosen, C.J. Bone As An Endocrine Organ. Endocr. Pr. 2012, 18, 758–762. [Google Scholar] [CrossRef]
  31. Xiao, W.; Wang, Y.; Pacios, S.; Li, S.; Graves, D.T. Cellular and Molecular Aspects of Bone Remodeling. Front. Oral. Biol. 2016, 18, 9–16. [Google Scholar] [CrossRef]
  32. Buck, D.W.; Dumanian, G.A. Bone Biology and Physiology. Plast. Reconstr. Surg. 2012, 129, 1314–1320. [Google Scholar] [CrossRef]
  33. Roeder, E.; Matthews, B.G.; Kalajzic, I. Visual reporters for study of the osteoblast lineage. Bone 2016, 92, 189–195. [Google Scholar] [CrossRef] [Green Version]
  34. Hou, Z.; Wang, Z.; Tao, Y.; Bai, J.; Yu, B.; Shen, J.; Sun, H.; Xiao, L.; Xu, Y.; Zhou, J.; et al. KLF2 regulates osteoblast differentiation by targeting of Runx2. Lab. Investig. 2019, 99, 271–280. [Google Scholar] [CrossRef]
  35. Gu, J.; Tong, X.-S.; Chen, G.-H.; Wang, D.; Chen, Y.; Yuan, Y.; Liu, X.-Z.; Bian, J.-C.; Liu, Z.-P. Effects of 1α,25-(OH)2D3 on the formation and activity of osteoclasts in RAW264.7 cells. J. Steroid Biochem. Mol. Biol. 2015, 152, 25–33. [Google Scholar] [CrossRef] [PubMed]
  36. Chiu, Y.-H.; Ritchlin, C.T. DC-STAMP: A Key Regulator in Osteoclast Differentiation. J. Cell. Physiol. 2016, 231, 2402–2407. [Google Scholar] [CrossRef]
  37. Chen, X.; Wang, Z.; Duan, N.; Zhu, G.; Schwarz, E.M.; Xie, C. Osteoblast–osteoclast interactions. Connect. Tissue Res. 2017, 59, 99–107. [Google Scholar] [CrossRef] [PubMed]
  38. Hemmatian, H.; Bakker, A.D.; Klein-Nulend, J.; Van Lenthe, G.H. Aging, Osteocytes, and Mechanotransduction. Curr. Osteoporos. Rep. 2017, 15, 401–411. [Google Scholar] [CrossRef] [PubMed]
  39. Tresguerres, F.; Torres, J.; López-Quiles, J.; Hernández, G.; Vega, J.; Tresguerres, I. The osteocyte: A multifunctional cell within the bone. Ann. Anat.-Anat. Anz. 2019, 227, 151422. [Google Scholar] [CrossRef]
  40. Shapses, S.A.; Pop, L.C.; Wang, Y. Obesity is a concern for bone health with aging. Nutr. Res. 2017, 39, 1–13. [Google Scholar] [CrossRef]
  41. Cao, J.J. Effects of obesity on bone metabolism. J. Orthop. Surg. Res. 2011, 6, 30. [Google Scholar] [CrossRef]
  42. Fintini, D.; Cianfarani, S.; Cofini, M.; Andreoletti, A.; Ubertini, G.M.; Cappa, M.; Manco, M. The Bones of Children with Obesity. Front. Endocrinol. 2020, 11, 200. [Google Scholar] [CrossRef]
  43. Marahleh, A.; Kitaura, H.; Ohori, F.; Kishikawa, A.; Ogawa, S.; Shen, W.-R.; Qi, J.; Noguchi, T.; Nara, Y.; Mizoguchi, I. TNF-α Directly Enhances Osteocyte RANKL Expression and Promotes Osteoclast Formation. Front. Immunol. 2019, 10, 2925. [Google Scholar] [CrossRef] [Green Version]
  44. Shinohara, H.; Teramachi, J.; Okamura, H.; Yang, D.; Nagata, T.; Haneji, T. Double Stranded RNA-Dependent Protein Kinase is Necessary for TNF-α-Induced Osteoclast Formation In Vitro and In Vivo. J. Cell. Biochem. 2015, 116, 1957–1967. [Google Scholar] [CrossRef] [PubMed]
  45. Luo, G.; Li, F.; Li, X.; Wang, Z.-G.; Zhang, B. TNF-α and RANKL promote osteoclastogenesis by upregulating RANK via the NF-κB pathway. Mol. Med. Rep. 2018, 17, 6605–6611. [Google Scholar] [CrossRef] [PubMed]
  46. Ohori, F.; Kitaura, H.; Ogawa, S.; Shen, W.-R.; Qi, J.; Noguchi, T.; Marahleh, A.; Nara, Y.; Pramusita, A.; Mizoguchi, I. IL-33 Inhibits TNF-α-Induced Osteoclastogenesis and Bone Resorption. Int. J. Mol. Sci. 2020, 21, 1130. [Google Scholar] [CrossRef] [PubMed]
  47. Shu, L.; Beier, E.; Sheu, T.; Zhang, H.; Zuscik, M.J.; Puzas, E.J.; Boyce, B.F.; Mooney, R.A.; Xing, L. High-Fat Diet Causes Bone Loss in Young Mice by Promoting Osteoclastogenesis Through Alteration of the Bone Marrow Environment. Calcif. Tissue Int. 2015, 96, 313–323. [Google Scholar] [CrossRef]
  48. Noguchi, T.; Kitaura, H.; Ogawa, S.; Qi, J.; Shen, W.-R.; Ohori, F.; Marahleh, A.; Nara, Y.; Pramusita, A.; Mizoguchi, I. TNF-α stimulates the expression of RANK during orthodontic tooth movement. Arch. Oral Biol. 2020, 117, 104796. [Google Scholar] [CrossRef]
  49. Min, H.-Y.; Son, H.-E.; Jang, W.-G. Alpha-pinene promotes osteoblast differentiation and attenuates TNFα-induced inhibition of differentiation in MC3T3-E1 pre-osteoblasts. Clin. Exp. Pharmacol. Physiol. 2019, 47, 831–837. [Google Scholar] [CrossRef]
  50. Abuna, R.P.; De Oliveira, F.S.; Santos, T.D.S.; Guerra, T.R.; Rosa, A.L.; Beloti, M.M. Participation of TNF-α in Inhibitory Effects of Adipocytes on Osteoblast Differentiation. J. Cell. Physiol. 2015, 231, 204–214. [Google Scholar] [CrossRef]
  51. Jeong, B.-C. ATF3 mediates the inhibitory action of TNF-α on osteoblast differentiation through the JNK signaling pathway. Biochem. Biophys. Res. Commun. 2018, 499, 696–701. [Google Scholar] [CrossRef]
  52. Wang, N.; Zhou, Z.; Wu, T.; Liu, W.; Yin, P.; Pan, C.; Yu, X. TNF-α-induced NF-κB activation upregulates microRNA-150-3p and inhibits osteogenesis of mesenchymal stem cells by targeting β-catenin. Open Biol. 2016, 6, 150258. [Google Scholar] [CrossRef]
  53. Ye, X.; Huang, H.; Zhao, N.; Zhang, J.; Yang, P. Inhibition of Runx2 signaling by TNF-α in ST2 murine bone marrow stromal cells undergoing osteogenic differentiation. Vitr. Cell. Dev. Biol.-Anim. 2016, 52, 1026–1033. [Google Scholar] [CrossRef]
  54. Bin, G.; Cuifang, W.; Bo, Z.; Jing, W.; Jin, J.; Xiaoyi, T.; Cong, C.; Yonggang, C.; Liping, A.; Jinglin, M.; et al. Fluid shear stress inhibits TNF-α-induced osteoblast apoptosis via ERK5 signaling pathway. Biochem. Biophys. Res. Commun. 2015, 466, 117–123. [Google Scholar] [CrossRef]
  55. Tsai, C.-L.; Chen, W.-C.; Hsieh, H.-L.; Chi, P.-L.; Hsiao, L.-D.; Yang, C.-M. TNF-α induces matrix metalloproteinase-9-dependent soluble intercellular adhesion molecule-1 release via TRAF2-mediated MAPKs and NF-κB activation in osteoblast-like MC3T3-E1 cells. J. Biomed. Sci. 2014, 21, 12. [Google Scholar] [CrossRef]
  56. Constanze, B.; Popper, B.; Aggarwal, B.B.; Shakibaei, M. Evidence that TNF-β suppresses osteoblast differentiation of mesenchymal stem cells and resveratrol reverses it through modulation of NF-κB, Sirt1 and Runx2. Cell Tissue Res. 2020, 381, 83–98. [Google Scholar] [CrossRef]
  57. Muluke, M.; Gold, T.; Kiefhaber, K.; Al-Sahli, A.; Celenti, R.; Jiang, H.; Cremers, S.; Van Dyke, T.; Schulze-Späte, U. Diet-Induced Obesity and Its Differential Impact on Periodontal Bone Loss. J. Dent. Res. 2015, 95, 223–229. [Google Scholar] [CrossRef]
  58. Zhang, K.; Wang, C.; Chen, Y.; Ji, X.; Chen, X.; Tian, L.; Yu, X. Preservation of high-fat diet-induced femoral trabecular bone loss through genetic target of TNF-α. Endocrine 2015, 50, 239–249. [Google Scholar] [CrossRef]
  59. Galozzi, P.; Bindoli, S.; Doria, A.; Sfriso, P. The revisited role of interleukin-1 alpha and beta in autoimmune and inflammatory disorders and in comorbidities. Autoimmun. Rev. 2021, 20, 102785. [Google Scholar] [CrossRef]
  60. Lee, J.; Park, C.; Kim, H.J.; Lee, Y.D.; Lee, Z.H.; Song, Y.W.; Kim, H.-H. Stimulation of osteoclast migration and bone resorption by C–C chemokine ligands 19 and 21. Exp. Mol. Med. 2017, 49, e358. [Google Scholar] [CrossRef]
  61. Cao, Y.; Jansen, I.D.C.; Sprangers, S.; Stap, J.; Leenen, P.J.; Everts, V.; de Vries, T.J. IL-1β differently stimulates proliferation and multinucleation of distinct mouse bone marrow osteoclast precursor subsets. J. Leukoc. Biol. 2016, 100, 513–523. [Google Scholar] [CrossRef]
  62. Son, H.S.; Lee, J.; Lee, H.I.; Kim, N.; Jo, Y.-J.; Lee, G.-R.; Hong, S.-E.; Kwon, M.; Kim, N.Y.; Kim, H.J.; et al. Benzydamine inhibits osteoclast differentiation and bone resorption via down-regulation of interleukin-1 expression. Acta Pharm. Sin. B 2019, 10, 462–474. [Google Scholar] [CrossRef]
  63. Wang, Y.; Galli, M.; Silver, A.S.; Lee, W.; Song, Y.; Mei, Y.M.; Bachus, C.; Glogauer, M.; McCulloch, C.A. IL1β and TNFα promote RANKL-dependent adseverin expression and osteoclastogenesis. J. Cell Sci. 2018, 131, jcs213967. [Google Scholar] [CrossRef] [Green Version]
  64. Lee, W.-S.; Lee, E.-G.; Sung, M.-S.; Choi, Y.-J.; Yoo, W.-H. Atorvastatin inhibits osteoclast differentiation by suppressing NF-κB and MAPK signaling during IL-1β-induced osteoclastogenesis. Korean J. Intern. Med. 2018, 33, 397–406. [Google Scholar] [CrossRef]
  65. Moon, S.-J.; Ahn, I.E.; Jung, H.; Yi, H.; Kim, J.; Kim, Y.; Kwok, S.-K.; Park, K.-S.; Min, J.-K.; Park, S.-H.; et al. Temporal differential effects of proinflammatory cytokines on osteoclastogenesis. Int. J. Mol. Med. 2013, 31, 769–777. [Google Scholar] [CrossRef]
  66. Qu, X.; Mei, J.; Yu, Z.; Zhai, Z.; Qiao, H.; Dai, K. Lenalidomide regulates osteocytes fate and related osteoclastogenesis via IL-1β/NF-κB/RANKL signaling. Biochem. Biophys. Res. Commun. 2018, 501, 547–555. [Google Scholar] [CrossRef]
  67. Ozeki, N.; Kawai, R.; Yamaguchi, H.; Hiyama, T.; Kinoshita, K.; Hase, N.; Nakata, K.; Kondo, A.; Mogi, M.; Nakamura, H. RETRACTED: IL-1β-induced matrix metalloproteinase-13 is activated by a disintegrin and metalloprotease-28-regulated proliferation of human osteoblast-like cells. Exp. Cell Res. 2014, 323, 165–177. [Google Scholar] [CrossRef]
  68. Hengartner, N.-E.; Fiedler, J.; Ignatius, A.; Brenner, R.E. IL-1β Inhibits Human Osteoblast Migration. Mol. Med. 2013, 19, 36–42. [Google Scholar] [CrossRef]
  69. Lazzerini, P.E.; Capperucci, C.; Spreafico, A.; Capecchi, P.L.; Niccolini, S.; Ferrata, P.; Frediani, B.; Galeazzi, M.; Laghi-Pasini, F. Rosuvastatin inhibits spontaneous and IL-1β-induced interleukin-6 production from human cultured osteoblastic cells. Jt. Bone Spine 2013, 80, 195–200. [Google Scholar] [CrossRef] [PubMed]
  70. Hah, Y.-S.; Kang, H.-G.; Cho, H.-Y.; Shin, S.-H.; Kim, U.-K.; Park, B.-W.; Lee, S.-I.; Rho, G.-J.; Kim, J.-R.; Byun, J.-H. JNK signaling plays an important role in the effects of TNF-α and IL-1β on in vitro osteoblastic differentiation of cultured human periosteal-derived cells. Mol. Biol. Rep. 2013, 40, 4869–4881. [Google Scholar] [CrossRef] [PubMed]
  71. Yang, H.; Liu, Q.; Ahn, J.H.; Kim, S.B.; Kim, Y.C.; Sung, S.H.; Hwang, B.Y.; Lee, M.K. Luteolin downregulates IL-1β-induced MMP-9 and -13 expressions in osteoblasts via inhibition of ERK signalling pathway. J. Enzym. Inhib. Med. Chem. 2011, 27, 261–266. [Google Scholar] [CrossRef] [PubMed]
  72. McKnight, Q.; Jenkins, S.; Li, X.; Nelson, T.; Marlier, A.; Cantley, L.G.; Finberg, K.E.; Fretz, J.A. IL-1β Drives Production of FGF-23 at the Onset of Chronic Kidney Disease in Mice. J. Bone Miner. Res. 2020, 35, 1352–1362. [Google Scholar] [CrossRef]
  73. Yamazaki, M.; Kawai, M.; Miyagawa, K.; Ohata, Y.; Tachikawa, K.; Kinoshita, S.; Nishino, J.; Ozono, K.; Michigami, T. Interleukin-1-induced acute bone resorption facilitates the secretion of fibroblast growth factor 23 into the circulation. J. Bone Miner. Metab. 2014, 33, 342–354. [Google Scholar] [CrossRef]
  74. Rupp, T.; Butscheidt, S.; Vettorazzi, E.; Oheim, R.; Barvencik, F.; Amling, M.; Rolvien, T. High FGF23 levels are associated with impaired trabecular bone microarchitecture in patients with osteoporosis. Osteoporos. Int. 2019, 30, 1655–1662. [Google Scholar] [CrossRef]
  75. He, Z.; Sun, Y.; Wu, J.; Xiong, Z.; Zhang, S.; Liu, J.; Liu, Y.; Li, H.; Jin, T.; Yang, Y.; et al. Evaluation of genetic variants in IL-1B and its interaction with the predisposition of osteoporosis in the northwestern Chinese Han population. J. Gene Med. 2020, 22, e3214. [Google Scholar] [CrossRef]
  76. Halade, G.V.; El Jamali, A.; Williams, P.J.; Fajardo, R.J.; Fernandes, G. Obesity-mediated inflammatory microenvironment stimulates osteoclastogenesis and bone loss in mice. Exp. Gerontol. 2011, 46, 43–52. [Google Scholar] [CrossRef]
  77. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2020, 33, 127–148. [Google Scholar] [CrossRef]
  78. Wu, Q.; Zhou, X.; Huang, D.; Ji, Y.; Kang, F. IL-6 Enhances Osteocyte-Mediated Osteoclastogenesis by Promoting JAK2 and RANKL Activity In Vitro. Cell. Physiol. Biochem. 2017, 41, 1360–1369. [Google Scholar] [CrossRef]
  79. Bakker, A.; Kulkarni, R.; Klein-Nulend, J.; Lems, W. IL-6 Alters Osteocyte Signaling toward Osteoblasts but Not Osteoclasts. J. Dent. Res. 2014, 93, 394–399. [Google Scholar] [CrossRef]
  80. McGregor, N.E.; Murat, M.; Elango, J.; Poulton, I.J.; Walker, E.C.; Crimeen-Irwin, B.; Ho, P.W.M.; Gooi, J.H.; Martin, T.J.; Sims, N.A. IL-6 exhibits both cis- and trans-signaling in osteocytes and osteoblasts, but only trans-signaling promotes bone formation and osteoclastogenesis. J. Biol. Chem. 2019, 294, 7850–7863. [Google Scholar] [CrossRef]
  81. He, B.; Yin, X.; Hao, D.; Zhang, X.; Zhang, Z.; Zhang, K.; Yang, X. Blockade of IL-6 alleviates bone loss induced by modeled microgravity in mice. Can. J. Physiol. Pharmacol. 2020, 98, 678–683. [Google Scholar] [CrossRef]
  82. Wang, C.; Tian, L.; Zhang, K.; Chen, Y.; Chen, X.; Xie, Y.; Zhao, Q.; Yu, X. Interleukin-6 gene knockout antagonizes high-fat-induced trabecular bone loss. J. Mol. Endocrinol. 2016, 57, 161–170. [Google Scholar] [CrossRef]
  83. Pathak, J.L.; Bakker, A.D.; Luyten, F.P.; Verschueren, P.; Lems, W.F.; Klein-Nulend, J.; Bravenboer, N. Systemic Inflammation Affects Human Osteocyte-Specific Protein and Cytokine Expression. Calcif. Tissue Int. 2016, 98, 596–608. [Google Scholar] [CrossRef]
  84. Yokota, K.; Sato, K.; Miyazaki, T.; Kitaura, H.; Kayama, H.; Miyoshi, F.; Araki, Y.; Akiyama, Y.; Takeda, K.; Mimura, T. Combination of Tumor Necrosis Factor α and Interleukin-6 Induces Mouse Osteoclast-like Cells with Bone Resorption Activity Both In Vitro and In Vivo. Arthritis Rheumatol. 2013, 66, 121–129. [Google Scholar] [CrossRef] [PubMed]
  85. Ricci, R.; Bevilacqua, F. The potential role of leptin and adiponectin in obesity: A comparative review. Veter. J. 2012, 191, 292–298. [Google Scholar] [CrossRef]
  86. Zhang, Y.; Chua, S., Jr. Leptin Function and Regulation. Compr. Physiol. 2017, 8, 351–369. [Google Scholar] [CrossRef] [PubMed]
  87. Obradovic, M.; Sudar-Milovanovic, E.; Soskic, S.; Essack, M.; Arya, S.; Stewart, A.J.; Gojobori, T.; Isenovic, E.R. Leptin and obesity: Role and clinical implication. Front. Endocrinol. 2021, 12, 585887. [Google Scholar] [CrossRef]
  88. Wang, Z.V.; Scherer, P.E. Adiponectin, the past two decades. J. Mol. Cell Biol. 2016, 8, 93–100. [Google Scholar] [CrossRef] [PubMed]
  89. Achari, A.E.; Jain, S.K. Adiponectin, a Therapeutic Target for Obesity, Diabetes, and Endothelial Dysfunction. Int. J. Mol. Sci. 2017, 18, 1321. [Google Scholar] [CrossRef]
  90. Upadhyay, J.; Farr, O.M.; Mantzoros, C.S. The role of leptin in regulating bone metabolism. Metabolism 2014, 64, 105–113. [Google Scholar] [CrossRef]
  91. China, S.P.; Sanyal, S.; Chattopadhyay, N. Adiponectin signaling and its role in bone metabolism. Cytokine 2018, 112, 116–131. [Google Scholar] [CrossRef]
  92. Guimarães, M.F.B.D.R.; de Andrade, M.V.M.; Machado, C.J.; Vieira, L.M.; Pinto, M.R.D.C.; Júnior, A.L.T.; Kakehasi, A.M. Leptin as an obesity marker in rheumatoid arthritis. Rheumatol. Int. 2018, 38, 1671–1677. [Google Scholar] [CrossRef]
  93. Fasshauer, M.; Blüher, M. Adipokines in health and disease. Trends Pharmacol. Sci. 2015, 36, 461–470. [Google Scholar] [CrossRef]
  94. Xie, C.; Chen, Q. Adipokines: New Therapeutic Target for Osteoarthritis? Curr. Rheumatol. Rep. 2019, 21, 71. [Google Scholar] [CrossRef]
  95. Yang, J.; Park, O.-J.; Kim, J.; Han, S.; Yang, Y.; Yun, C.-H.; Han, S.H. Adiponectin Deficiency Triggers Bone Loss by Up-Regulation of Osteoclastogenesis and Down-Regulation of Osteoblastogenesis. Front. Endocrinol. 2019, 10, 815. [Google Scholar] [CrossRef]
  96. Wang, F.; Wang, P.-X.; Wu, X.-L.; Dang, S.-Y.; Chen, Y.; Ni, Y.-Y.; Gao, L.-H.; Lu, S.-Y.; Kuang, Y.; Huang, L.; et al. Deficiency of Adiponectin Protects against Ovariectomy-Induced Osteoporosis in Mice. PLoS ONE 2013, 8, e68497. [Google Scholar] [CrossRef] [PubMed]
  97. Tu, Q.; Zhang, J.; Dong, L.Q.; Saunders, E.; Luo, E.; Tang, J.; Chen, J. Adiponectin Inhibits Osteoclastogenesis and Bone Resorption via APPL1-mediated Suppression of Akt1. J. Biol. Chem. 2011, 286, 12542–12553. [Google Scholar] [CrossRef]
  98. Abbott, M.J.; Roth, T.M.; Ho, L.; Wang, L.; O’Carroll, D.; Nissenson, R.A. Negative Skeletal Effects of Locally Produced Adiponectin. PLoS ONE 2015, 10, e0134290. [Google Scholar] [CrossRef]
  99. Liu, Z.; Liu, H.; Li, Y.; Wang, Y.; Xing, R.; Mi, F.; Xiang, C.; Fu, R. Adiponectin inhibits the differentiation and maturation of osteoclasts via the mTOR pathway in multiple myeloma. Int. J. Mol. Med. 2020, 45, 1112–1120. [Google Scholar] [CrossRef] [PubMed]
  100. Chen, G.; Huang, L.; Wu, X.; Liu, X.; Xu, Q.; Li, F.; Dai, M.; Zhang, B. Adiponectin inhibits osteoclastogenesis by suppressing NF-κB and p38 signaling pathways. Biochem. Biophys. Res. Commun. 2018, 503, 2075–2082. [Google Scholar] [CrossRef] [PubMed]
  101. Wu, X.; Huang, L.; Liu, J. Effects of adiponectin on osteoclastogenesis from mouse bone marrow-derived monocytes. Exp. Ther. Med. 2018, 17, 1228–1233. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, H.; Liu, S.; Ji, H.; Zhao, Q.; Liu, Y.; Hu, P.; Luo, E. An adiponectin receptor agonist promote osteogenesis via regulating bone-fat balance. Cell Prolif. 2021, 54, e13035. [Google Scholar] [CrossRef] [PubMed]
  103. China, S.P.; Pal, S.; Chattopadhyay, S.; Porwal, K.; Kushwaha, S.; Bhattacharyya, S.; Mittal, M.; Gurjar, A.A.; Barbhuyan, T.; Singh, A.K.; et al. Globular adiponectin reverses osteo-sarcopenia and altered body composition in ovariectomized rats. Bone 2017, 105, 75–86. [Google Scholar] [CrossRef]
  104. Haugen, S.; He, J.; Sundaresan, A.; Stunes, A.K.; Aasarød, K.M.; Tiainen, H.; Syversen, U.; Skallerud, B.; Reseland, J.E. Adiponectin Reduces Bone Stiffness: Verified in a Three-Dimensional Artificial Human Bone Model In Vitro. Front. Endocrinol. 2018, 9, 236. [Google Scholar] [CrossRef] [PubMed]
  105. Pacheco-Pantoja, E.L.; Fraser, W.D.; Wilson, P.J.M.; Gallagher, J.A. Differential effects of adiponectin in osteoblast-like cells. J. Recept. Signal Transduct. 2014, 34, 351–360. [Google Scholar] [CrossRef] [PubMed]
  106. Yin, G.; Liu, H.; Li, J.; Liu, Y.; Liu, X.; Luo, E. Adenoviral delivery of adiponectin ameliorates osteogenesis around implants in ovariectomized rats. J. Gene Med. 2019, 21, e3069. [Google Scholar] [CrossRef] [PubMed]
  107. Dragojevič, J.; Zupan, J.; Haring, G.; Herman, S.; Komadina, R.; Marc, J. Triglyceride metabolism in bone tissue is associated with osteoblast and osteoclast differentiation: A gene expression study. J. Bone Miner. Metab. 2013, 31, 512–519. [Google Scholar] [CrossRef]
  108. Yue, R.; Zhou, B.O.; Shimada, I.S.; Zhao, Z.; Morrison, S.J. Leptin Receptor Promotes Adipogenesis and Reduces Osteogenesis by Regulating Mesenchymal Stromal Cells in Adult Bone Marrow. Cell Stem Cell 2016, 18, 782–796. [Google Scholar] [CrossRef]
  109. Li, W.; Xu, P.; Wang, C.; Ha, X.; Gu, Y.; Wang, Y.; Zhang, J.; Xie, J. The effects of fat-induced obesity on bone metabolism in rats. Obes. Res. Clin. Pr. 2017, 11, 454–463. [Google Scholar] [CrossRef]
  110. Carnovali, M.; Luzi, L.; Terruzzi, I.; Banfi, G.; Mariotti, M. Metabolic and bone effects of high-fat diet in adult zebrafish. Endocrine 2017, 61, 317–326. [Google Scholar] [CrossRef]
  111. Feresin, R.G.; Johnson, S.A.; Elam, M.L.; Jo, E.; Arjmandi, B.H.; Hakkak, R. Effects of Obesity on Bone Mass and Quality in Ovariectomized Female Zucker Rats. J. Obes. 2014, 2014, 690123. [Google Scholar] [CrossRef]
  112. Philbrick, K.A.; Wong, C.P.; Branscum, A.J.; Turner, R.T.; Iwaniec, U.T. Leptin stimulates bone formation in ob/ob mice at doses having minimal impact on energy metabolism. J. Endocrinol. 2017, 232, 461–474. [Google Scholar] [CrossRef]
  113. Wu, Z.; Shao, P.; Dass, C.R.; Wei, Y. Systemic leptin administration alters callus VEGF levels and enhances bone fracture healing in wildtype and ob/ob mice. Injury 2018, 49, 1739–1745. [Google Scholar] [CrossRef]
  114. Tencerova, M.; Frost, M.; Figeac, F.; Nielsen, T.K.; Ali, D.; Lauterlein, J.-J.L.; Andersen, T.L.; Haakonsson, A.K.; Rauch, A.; Madsen, J.S.; et al. Obesity-Associated Hypermetabolism and Accelerated Senescence of Bone Marrow Stromal Stem Cells Suggest a Potential Mechanism for Bone Fragility. Cell Rep. 2019, 27, 2050–2062.e6. [Google Scholar] [CrossRef]
  115. Li, J.; Gao, Y.; Yu, T.; Lange, J.K.; LeBoff, M.S.; Gorska, A.; Luu, S.; Zhou, S.; Glowacki, J. Obesity and leptin influence vitamin D metabolism and action in human marrow stromal cells. J. Steroid Biochem. Mol. Biol. 2019, 198, 105564. [Google Scholar] [CrossRef]
  116. Maggio, A.B.; Belli, D.C.; Puigdefabregas, J.W.B.; Rizzoli, R.; Farpour-Lambert, N.J.; Beghetti, M.; McLin, V.A. High Bone Density in Adolescents with Obesity Is Related to Fat Mass and Serum Leptin Concentrations. J. Craniofacial Surg. 2014, 58, 723–728. [Google Scholar] [CrossRef]
  117. Nakamura, Y.; Nakano, M.; Suzuki, T.; Sato, J.; Kato, H.; Takahashi, J.; Shiraki, M. Two adipocytokines, leptin and adiponectin, independently predict osteoporotic fracture risk at different bone sites in postmenopausal women. Bone 2020, 137, 115404. [Google Scholar] [CrossRef] [PubMed]
  118. Dimitri, P.; Jacques, R.; Paggiosi, M.; King, D.; Walsh, J.; Taylor, Z.; Frangi, A.; Bishop, N.; Eastell, R. Leptin May Play a Role in Bone Microstructural Alterations in Obese Children. J. Clin. Endocrinol. Metab. 2015, 100, 594–602. [Google Scholar] [CrossRef]
  119. Reid, I.R.; A Baldock, P.; Cornish, J. Effects of Leptin on the Skeleton. Endocr. Rev. 2018, 39, 938–959. [Google Scholar] [CrossRef] [PubMed]
  120. Wong, I.P.; Nguyen, A.D.; Khor, E.C.; Enriquez, R.F.; A Eisman, J.; Sainsbury, A.; Herzog, H.; A Baldock, P. Neuropeptide Y is a critical modulator of Leptin’s regulation of cortical bone. J. Bone Miner. Res. 2012, 28, 886–898. [Google Scholar] [CrossRef]
  121. Wee, N.K.Y.; Enriquez, R.F.; Nguyen, A.; Horsnell, H.; Kulkarni, R.; Khor, E.C.; Herzog, H.; Baldock, P.A. Diet-induced obesity suppresses cortical bone accrual by a neuropeptide Y-dependent mechanism. Int. J. Obes. 2018, 42, 1925–1938. [Google Scholar] [CrossRef]
  122. Srivastava, G.; Apovian, C. Future Pharmacotherapy for Obesity: New Anti-obesity Drugs on the Horizon. Curr. Obes. Rep. 2018, 7, 147–161. [Google Scholar] [CrossRef]
  123. Erener, T.; Ceritoğlu, K.U.; Aktekin, C.N.; Dalgic, A.D.; Keskin, D.; Geneci, F.; Ocak, M.; Bilecenoğlu, B.; Hücümenoğlu, S.; Çaydere, M.; et al. Investigation of the effect of ghrelin on bone fracture healing in rats. Clin. Exp. Pharmacol. Physiol. 2021, 48, 1382–1390. [Google Scholar] [CrossRef] [PubMed]
  124. Xiao, L.; Zhang, H.; Wang, Y.; Li, J.; Yang, G.; Wang, L.; Liang, Z. Dysregulation of the ghrelin/RANKL/OPG pathway in bone mass is related to AIS osteopenia. Bone 2020, 134, 115291. [Google Scholar] [CrossRef] [PubMed]
  125. Müller, T.; Finan, B.; Bloom, S.; D’Alessio, D.; Drucker, D.; Flatt, P.; Fritsche, A.; Gribble, F.; Grill, H.; Habener, J.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef] [PubMed]
  126. Meng, J.; Ma, X.; Wang, N.; Jia, M.; Bi, L.; Wang, Y.; Li, M.; Zhang, H.; Xue, X.; Hou, Z.; et al. Activation of GLP-1 Receptor Promotes Bone Marrow Stromal Cell Osteogenic Differentiation through β-Catenin. Stem Cell Rep. 2016, 6, 579–591. [Google Scholar] [CrossRef] [PubMed]
  127. Mansur, S.A.; Mieczkowska, A.; Flatt, P.R.; Chappard, D.; Irwin, N.; Mabilleau, G. The GLP-1 Receptor Agonist Exenatide Ameliorates Bone Composition and Tissue Material Properties in High Fat Fed Diabetic Mice. Front. Endocrinol. 2019, 10, 51. [Google Scholar] [CrossRef]
  128. Kirk, B.; Feehan, J.; Lombardi, G.; Duque, G. Muscle, Bone, and Fat Crosstalk: The Biological Role of Myokines, Osteokines, and Adipokines. Curr. Osteoporos. Rep. 2020, 18, 388–400. [Google Scholar] [CrossRef]
  129. Ilich, J.Z.; Gilman, J.C.; Cvijetic, S.; Boschiero, D. Chronic Stress Contributes to Osteosarcopenic Adiposity via Inflammation and Immune Modulation: The Case for More Precise Nutritional Investigation. Nutrients 2020, 12, 989. [Google Scholar] [CrossRef]
  130. Al Saedi, A.; Hassan, E.B.; Duque, G. The diagnostic role of fat in osteosarcopenia. J. Lab. Precis. Med. 2019, 4, 7. [Google Scholar] [CrossRef]
  131. Li, H.; Wang, F.; Yang, M.; Sun, J.; Zhao, Y.; Tang, D. The Effect of Irisin as a Metabolic Regulator and Its Therapeutic Potential for Obesity. Int. J. Endocrinol. 2021, 2021, 6572342. [Google Scholar] [CrossRef]
  132. Xue, Y.; Hu, S.; Chen, C.; He, J.; Sun, J.; Jin, Y.; Zhang, Y.; Zhu, G.; Shi, Q.; Rui, Y. Myokine Irisin promotes osteogenesis by activating BMP/SMAD signaling via αV integrin and regulates bone mass in mice. Int. J. Biol. Sci. 2022, 18, 572–584. [Google Scholar] [CrossRef]
  133. He, X.; Hua, Y.; Li, Q.; Zhu, W.; Pan, Y.; Yang, Y.; Li, X.; Wu, M.; Wang, J.; Gan, X. FNDC5/irisin facilitates muscle−adipose−bone connectivity through ubiquitination-dependent activation of runt-related transcriptional factors RUNX1/2. J. Biol. Chem. 2022, 298, 101679. [Google Scholar] [CrossRef]
  134. Ozgor, B.Y.; Demiral, I.; Zeybek, U.; Celik, F.; Buyru, F.; Yeh, J.; Bastu, E. Effects of Irisin Compared with Exercise on Specific Metabolic and Obesity Parameters in Female Mice with Obesity. Metab. Syndr. Relat. Disord. 2020, 18, 141–145. [Google Scholar] [CrossRef]
  135. Kang, Y.S.; Kim, J.C.; Kim, J.S.; Kim, S.H. Effects of Swimming Exercise on Serum Irisin and Bone FNDC5 in Rat Models of High-Fat Diet-Induced Osteoporosis. J. Sport. Sci. Med. 2019, 18, 596–603. [Google Scholar]
  136. Bäckhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The Gut Microbiota as an Environmental Factor That Regulates Fat Storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef] [PubMed]
  137. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An Obesity-Associated Gut Microbiome with Increased Capacity for Energy Harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  138. Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
  139. Liu, H.; Wang, P.; Cao, M.; Li, M.; Wang, F. Protective Role of Oligomycin Against Intestinal Epithelial Barrier Dysfunction Caused by IFN-? and TNF-a. Cell. Physiol. Biochem. 2012, 29, 799–808. [Google Scholar] [CrossRef] [PubMed]
  140. Saad, M.J.A.; Santos, A.; Prada, P.O. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology 2016, 31, 283–293. [Google Scholar] [CrossRef] [PubMed]
  141. Luo, Y.; Chen, G.-L.; Hannemann, N.; Ipseiz, N.; Krönke, G.; Bäuerle, T.; Munos, L.; Wirtz, S.; Schett, G.; Bozec, A. Microbiota from Obese Mice Regulate Hematopoietic Stem Cell Differentiation by Altering the Bone Niche. Cell Metab. 2015, 22, 886–894. [Google Scholar] [CrossRef]
  142. Sato, K.; Yamazaki, K.; Kato, T.; Nakanishi, Y.; Tsuzuno, T.; Yokoji-Takeuchi, M.; Yamada-Hara, M.; Miura, N.; Okuda, S.; Ohno, H.; et al. Obesity-Related Gut Microbiota Aggravates Alveolar Bone Destruction in Experimental Periodontitis through Elevation of Uric Acid. Mbio 2021, 12, e0077121. [Google Scholar] [CrossRef]
  143. Gomes, A.C.; Hoffmann, C.; Mota, J.F. The human gut microbiota: Metabolism and perspective in obesity. Gut Microbes 2018, 9, 308–325. [Google Scholar] [CrossRef]
  144. Stojanov, S.; Berlec, A.; Štrukelj, B. The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio in the Treatment of Obesity and Inflammatory Bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef] [PubMed]
  145. McCabe, L.R.; Irwin, R.; Tekalur, A.; Evans, C.; Schepper, J.D.; Parameswaran, N.; Ciancio, M. Exercise prevents high fat diet-induced bone loss, marrow adiposity and dysbiosis in male mice. Bone 2018, 118, 20–31. [Google Scholar] [CrossRef] [PubMed]
  146. Xie, W.; Han, Y.; Li, F.; Gu, X.; Su, D.; Yu, W.; Li, Z.; Xiao, J. Neuropeptide Y1 Receptor Antagonist Alters Gut Microbiota and Alleviates the Ovariectomy-Induced Osteoporosis in Rats. Calcif. Tissue Int. 2019, 106, 444–454. [Google Scholar] [CrossRef] [PubMed]
  147. Méndez-Salazar, E.O.; Ortiz-López, M.G.; Granados-Silvestre, M.D.L.; Palacios-González, B.; Menjivar, M. Altered Gut Microbiota and Compositional Changes in Firmicutes and Proteobacteria in Mexican Undernourished and Obese Children. Front. Microbiol. 2018, 9, 2494. [Google Scholar] [CrossRef] [PubMed]
  148. Palacios-González, B.; Ramírez-Salazar, E.G.; Rivera-Paredez, B.; Quiterio, M.; Flores, Y.N.; Macias-Kauffer, L.; Moran-Ramos, S.; Denova-Gutiérrez, E.; Ibarra-González, I.; Vela-Amieva, M.; et al. A Multi-Omic Analysis for Low Bone Mineral Density in Postmenopausal Women Suggests a Relationship between Diet, Metabolites, and Microbiota. Microorganisms 2020, 8, 1630. [Google Scholar] [CrossRef] [PubMed]
  149. Amabebe, E.; Robert, F.O.; Agbalalah, T.; Orubu, E.S.F. Microbial dysbiosis-induced obesity: Role of gut microbiota in homoeostasis of energy metabolism. Br. J. Nutr. 2020, 123, 1127–1137. [Google Scholar] [CrossRef] [PubMed]
  150. Liu, J.; Chen, C.; Liu, Z.; Luo, Z.; Rao, S.; Jin, L.; Wan, T.; Yue, T.; Tan, Y.; Yin, H.; et al. Extracellular Vesicles from Child Gut Microbiota Enter into Bone to Preserve Bone Mass and Strength. Adv. Sci. 2021, 8, 2004831. [Google Scholar] [CrossRef]
  151. Huck, O.; Mulhall, H.; Rubin, G.; Kizelnik, Z.; Iyer, R.; Perpich, J.D.; Haque, N.; Cani, P.D.; de Vos, W.M.; Amar, S. Akkermansia muciniphila reduces Porphyromonas gingivalis -induced inflammation and periodontal bone destruction. J. Clin. Periodontol. 2019, 47, 202–212. [Google Scholar] [CrossRef] [PubMed]
  152. Raftar, S.K.A.; Tavassol, Z.H.; Amiri, M.; Ejtahed, H.-S.; Zangeneh, M.; Sadeghi, S.; Ashrafian, F.; Kariman, A.; Khatami, S.; Siadat, S.D. Assessment of fecal Akkermansia muciniphila in patients with osteoporosis and osteopenia: A pilot study. J. Diabetes Metab. Disord. 2021, 20, 279–284. [Google Scholar] [CrossRef]
  153. Fernández-Murga, M.L.; Olivares, M.; Sanz, Y. Bifidobacterium pseudocatenulatum CECT 7765 reverses the adverse effects of diet-induced obesity through the gut-bone axis. Bone 2020, 141, 115580. [Google Scholar] [CrossRef]
  154. Behera, J.; Ison, J.; Voor, M.J.; Tyagi, N. Probiotics Stimulate Bone Formation in Obese Mice via Histone Methylations. Theranostics 2021, 11, 8605–8623. [Google Scholar] [CrossRef] [PubMed]
  155. Eaimworawuthikul, S.; Tunapong, W.; Chunchai, T.; Yasom, S.; Wanchai, K.; Suntornsaratoon, P.; Charoenphandhu, N.; Thiennimitr, P.; Chattipakorn, N.; Chattipakorn, S.C. Effects of probiotics, prebiotics or synbiotics on jawbone in obese-insulin resistant rats. Eur. J. Nutr. 2018, 58, 2801–2810. [Google Scholar] [CrossRef] [PubMed]
  156. Eaimworawuthikul, S.; Tunapong, W.; Chunchai, T.; Suntornsaratoon, P.; Charoenphandhu, N.; Thiennimitr, P.; Chattipakorn, N.; Chattipakorn, S.C. Altered gut microbiota ameliorates bone pathology in the mandible of obese–insulin-resistant rats. Eur. J. Nutr. 2019, 59, 1453–1462. [Google Scholar] [CrossRef] [PubMed]
  157. Tyagi, A.M.; Yu, M.; Darby, T.M.; Vaccaro, C.; Li, J.-Y.; Owens, J.A.; Hsu, E.; Adams, J.; Weitzmann, M.N.; Jones, R.M.; et al. The Microbial Metabolite Butyrate Stimulates Bone Formation via T Regulatory Cell-Mediated Regulation of WNT10B Expression. Immunity 2018, 49, 1116–1131.e7. [Google Scholar] [CrossRef] [PubMed]
  158. Sun, X.; Zhao, H.; Liu, Z.; Sun, X.; Zhang, D.; Wang, S.; Xu, Y.; Zhang, G.; Wang, D. Modulation of Gut Microbiota by Fucoxanthin During Alleviation of Obesity in High-Fat Diet-Fed Mice. J. Agric. Food Chem. 2020, 68, 5118–5128. [Google Scholar] [CrossRef] [PubMed]
  159. Zhang, Z.; Lin, T.; Meng, Y.; Hu, M.; Shu, L.; Jiang, H.; Gao, R.; Ma, J.; Wang, C.; Zhou, X. FOS/GOS attenuates high-fat diet induced bone loss via reversing microbiota dysbiosis, high intestinal permeability and systemic inflammation in mice. Metabolism 2021, 119, 154767. [Google Scholar] [CrossRef] [PubMed]
  160. Rios, J.L.; Bomhof, M.R.; Reimer, R.A.; Hart, D.A.; Collins, K.H.; Herzog, W. Protective effect of prebiotic and exercise intervention on knee health in a rat model of diet-induced obesity. Sci. Rep. 2019, 9, 3893. [Google Scholar] [CrossRef]
  161. Kim, K.-J.; Lee, J.; Wang, W.; Lee, Y.; Oh, E.; Park, K.-H.; Park, C.; Woo, G.-E.; Son, Y.-J.; Kang, H. Austalide K from the Fungus Penicillium rudallense Prevents LPS-Induced Bone Loss in Mice by Inhibiting Osteoclast Differentiation and Promoting Osteoblast Differentiation. Int. J. Mol. Sci. 2021, 22, 5493. [Google Scholar] [CrossRef]
  162. Goel, P.N.; Egol, A.J.; Moharrer, Y.; Brandfield-Harvey, B.; Ahn, J.; Ashley, J.W. Notch signaling inhibition protects against LPS mediated osteolysis. Biochem. Biophys. Res. Commun. 2019, 515, 538–543. [Google Scholar] [CrossRef]
  163. Kishikawa, A.; Kitaura, H.; Kimura, K.; Ogawa, S.; Qi, J.; Shen, W.-R.; Ohori, F.; Noguchi, T.; Marahleh, A.; Nara, Y.; et al. Docosahexaenoic Acid Inhibits Inflammation-Induced Osteoclast Formation and Bone Resorption in vivo Through GPR120 by Inhibiting TNF-α Production in Macrophages and Directly Inhibiting Osteoclast Formation. Front. Endocrinol. 2019, 10, 157. [Google Scholar] [CrossRef]
  164. Jin, J.; Machado, E.R.; Yu, H.; Zhang, X.; Lu, Z.; Li, Y.; Lopes-Virella, M.F.; Kirkwood, K.; Huang, Y. Simvastatin inhibits LPS-induced alveolar bone loss during metabolic syndrome. J. Dent. Res. 2013, 93, 294–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Guo, C.; Yuan, L.; Wang, J.-G.; Wang, F.; Yang, X.-K.; Zhang, F.-H.; Song, J.-L.; Ma, X.-Y.; Cheng, Q.; Song, G.-H. Lipopolysaccharide (LPS) Induces the Apoptosis and Inhibits Osteoblast Differentiation Through JNK Pathway in MC3T3-E1 Cells. Inflammation 2013, 37, 621–631. [Google Scholar] [CrossRef] [PubMed]
  166. Gao, A.; Wang, X.; Yu, H.; Li, N.; Hou, Y.; Yu, W. Effect of Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) on the expression of EphA2 in osteoblasts and osteoclasts. Vitr. Cell. Dev. Biol.-Anim. 2015, 52, 228–234. [Google Scholar] [CrossRef] [PubMed]
  167. Chang, Y.; Hu, C.-C.; Wu, Y.-Y.; Ueng, S.; Chang, C.-H.; Chen, M.-F. Ibudilast Mitigates Delayed Bone Healing Caused by Lipopolysaccharide by Altering Osteoblast and Osteoclast Activity. Int. J. Mol. Sci. 2021, 22, 1169. [Google Scholar] [CrossRef] [PubMed]
  168. Irie, N.; Takada, Y.; Watanabe, Y.; Matsuzaki, Y.; Naruse, C.; Asano, M.; Iwakura, Y.; Suda, T.; Matsuo, K. Bidirectional Signaling through EphrinA2-EphA2 Enhances Osteoclastogenesis and Suppresses Osteoblastogenesis. J. Biol. Chem. 2009, 284, 14637–14644. [Google Scholar] [CrossRef]
  169. Xing, Q.; de Vos, P.; Faas, M.; Ye, Q.; Ren, Y. LPS Promotes Pre-osteoclast Activity by Up-regulating CXCR4 via TLR-4. J. Dent. Res. 2011, 90, 157–162. [Google Scholar] [CrossRef]
  170. Han, Y.; You, X.; Xing, W.; Zhang, Z.; Zou, W. Paracrine and endocrine actions of bone—the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res. 2018, 6, 16. [Google Scholar] [CrossRef]
  171. Ho, B.B.; Bergwitz, C. FGF23 signalling and physiology. J. Mol. Endocrinol. 2021, 66, R23–R32. [Google Scholar] [CrossRef]
  172. Prié, D. FGF23 and Cardiovascular Risk. Ann. d’Endocrinologie 2020, 82, 141–143. [Google Scholar] [CrossRef]
  173. Mizokami, A.; Kawakubo-Yasukochi, T.; Hirata, M. Osteocalcin and its endocrine functions. Biochem. Pharmacol. 2017, 132, 1–8. [Google Scholar] [CrossRef]
  174. Mosialou, I.; Shikhel, S.; Liu, J.-M.; Maurizi, A.; Luo, N.; He, Z.; Huang, Y.; Zong, H.; Friedman, R.A.; Barasch, J.; et al. MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Nature 2017, 543, 385–390. [Google Scholar] [CrossRef] [PubMed]
  175. Yaylali, Y.T.; Fidan-Yaylali, G.; Dedeoglu, O.; Senol, H. Osteocalcin and epicardial adipose tissue in obesity: New hints for epicardial adipose tissue–bone crosstalk. Scand. Cardiovasc. J. 2019, 53, 296–298. [Google Scholar] [CrossRef] [PubMed]
  176. Geserick, M.; Vogel, M.; Eckelt, F.; Schlingmann, M.; Hiemisch, A.; Baber, R.; Thiery, J.; Körner, A.; Kiess, W.; Kratzsch, J. Children and adolescents with obesity have reduced serum bone turnover markers and 25-hydroxyvitamin D but increased parathyroid hormone concentrations—Results derived from new pediatric reference ranges. Bone 2019, 132, 115124. [Google Scholar] [CrossRef] [PubMed]
  177. Kim, G.S.; Jekal, Y.; Kim, H.S.; Im, J.-A.; Park, J.Y.; Chu, S.H. Reduced serum total osteocalcin is associated with central obesity in Korean children. Obes. Res. Clin. Pr. 2014, 8, e230–e237. [Google Scholar] [CrossRef] [PubMed]
  178. Razny, U.; Goralska, J.; Zdzienicka, A.; Fedak, D.; Masania, J.; Rabbani, N.; Thornalley, P.; Pawlica-Gosiewska, D.; Gawlik, K.; Dembinska-Kiec, A.; et al. Relation of the protein glycation, oxidation and nitration to the osteocalcin level in obese subjects. Acta Biochim. Pol. 2017, 64, 415–422. [Google Scholar] [CrossRef] [PubMed]
  179. Vigevano, F.; Gregori, G.; Colleluori, G.; Chen, R.; Autemrongsawat, V.; Napoli, N.; Qualls, C.; Villareal, D.T.; Armamento-Villareal, R. In Men With Obesity, T2DM Is Associated with Poor Trabecular Microarchitecture and Bone Strength and Low Bone Turnover. J. Clin. Endocrinol. Metab. 2021, 106, 1362–1376. [Google Scholar] [CrossRef]
  180. Guedes, J.; Esteves, J.; Morais, M.; Zorn, T.; Furuya, D. Osteocalcin improves insulin resistance and inflammation in obese mice: Participation of white adipose tissue and bone. Bone 2018, 115, 68–82. [Google Scholar] [CrossRef]
  181. Zhou, B.; Li, H.; Liu, J.; Xu, L.; Guo, Q.; Zang, W.; Sun, H.; Wu, S. Autophagic dysfunction is improved by intermittent administration of osteocalcin in obese mice. Int. J. Obes. 2016, 40, 833–843. [Google Scholar] [CrossRef]
  182. Ferron, M.; McKee, M.D.; Levine, R.L.; Ducy, P.; Karsenty, G. Intermittent injections of osteocalcin improve glucose metabolism and prevent type 2 diabetes in mice. Bone 2012, 50, 568–575. [Google Scholar] [CrossRef]
  183. Hanks, L.J.; Casazza, K.; Judd, S.E.; Jenny, N.S.; Gutiérrez, O.M. Associations of Fibroblast Growth Factor-23 with Markers of Inflammation, Insulin Resistance and Obesity in Adults. PLoS ONE 2015, 10, e0122885. [Google Scholar] [CrossRef]
  184. Zaheer, S.; De Boer, I.H.; Allison, M.; Brown, J.M.; Psaty, B.M.; Robinson-Cohen, C.; Michos, E.D.; Ix, J.H.; Kestenbaum, B.; Siscovick, D.; et al. Fibroblast Growth Factor 23, Mineral Metabolism, and Adiposity in Normal Kidney Function. J. Clin. Endocrinol. Metab. 2017, 102, 1387–1395. [Google Scholar] [CrossRef] [PubMed]
  185. Hu, X.; Ma, X.; Luo, Y.; Xu, Y.; Xiong, Q.; Pan, X.; Xiao, Y.; Bao, Y.; Jia, W. Associations of serum fibroblast growth factor 23 levels with obesity and visceral fat accumulation. Clin. Nutr. 2016, 37, 223–228. [Google Scholar] [CrossRef] [PubMed]
  186. Grethen, E.; Hill, K.M.; Jones, R.; Cacucci, B.M.; Gupta, C.E.; Acton, A.; Considine, R.V.; Peacock, M. Serum Leptin, Parathyroid Hormone, 1,25-Dihydroxyvitamin D, Fibroblast Growth Factor 23, Bone Alkaline Phosphatase, and Sclerostin Relationships in Obesity. J. Clin. Endocrinol. Metab. 2012, 97, 1655–1662. [Google Scholar] [CrossRef] [PubMed]
  187. Xu, L.; Zhang, L.; Zhang, H.; Yang, Z.; Qi, L.; Wang, Y.; Ren, S. The participation of fibroblast growth factor 23 (FGF23) in the progression of osteoporosis via JAK/STAT pathway. J. Cell. Biochem. 2017, 119, 3819–3828. [Google Scholar] [CrossRef]
  188. Garland, J.S.; Holden, R.M.; Ross, R.; Adams, M.A.; Nolan, R.; Hopman, W.M.; Morton, A.R. Insulin resistance is associated with Fibroblast Growth Factor-23 in stage 3–5 chronic kidney disease patients. J. Diabetes Its Complicat. 2013, 28, 61–65. [Google Scholar] [CrossRef]
  189. Saito, M.; Marumo, K. Bone Quality in Diabetes. Front. Endocrinol. 2013, 4, 72. [Google Scholar] [CrossRef]
  190. Liu, C.; Jiang, D. High glucose-induced LIF suppresses osteoblast differentiation via regulating STAT3/SOCS3 signaling. Cytokine 2017, 91, 132–139. [Google Scholar] [CrossRef]
  191. Yang, L.; Liu, J.; Shan, Q.; Geng, G.; Shao, P. High glucose inhibits proliferation and differentiation of osteoblast in alveolar bone by inducing pyroptosis. Biochem. Biophys. Res. Commun. 2019, 522, 471–478. [Google Scholar] [CrossRef]
  192. Bornstein, S.; Moschetta, M.; Kawano, Y.; Sacco, A.; Huynh, D.; Brooks, D.; Manier, S.; Fairfield, H.; Falank, C.; Roccaro, A.M.; et al. Metformin Affects Cortical Bone Mass and Marrow Adiposity in Diet-Induced Obesity in Male Mice. Endocrinology 2017, 158, 3369–3385. [Google Scholar] [CrossRef]
  193. Qu, B.; Gong, K.; Yang, H.; Li, Y.; Jiang, T.; Zeng, Z.; Cao, Z.; Pan, X. SIRT1 suppresses high glucose and palmitate-induced osteoclast differentiation via deacetylating p66Shc. Mol. Cell. Endocrinol. 2018, 474, 97–104. [Google Scholar] [CrossRef]
  194. Xu, F.; Ye, Y.-P.; Dong, Y.-H.; Guo, F.-J.; Chen, A.-M.; Huang, S.-L. Inhibitory effects of high glucose/insulin environment on osteoclast formation and resorption in vitro. J. Huazhong Univ. Sci. Technol. 2013, 33, 244–249. [Google Scholar] [CrossRef] [PubMed]
  195. Mosialou, I.; Shikhel, S.; Luo, N.; Petropoulou, P.I.; Panitsas, K.; Bisikirska, B.; Rothman, N.J.; Tenta, R.; Cariou, B.; Wargny, M.; et al. Lipocalin-2 counteracts metabolic dysregulation in obesity and diabetes. J. Exp. Med. 2020, 217, e20191261. [Google Scholar] [CrossRef] [PubMed]
  196. Wen, X.; Zhu, B.; Zhang, Y.; Mei, F.; Cheng, X.; Qian, C.; Yang, P.; Lu, L.; Zhou, D.; Gao, J.; et al. Alterations in Fat Mass and Bone Mineral Density Are Associated with Decreased Lipocalin-2 After Laparoscopic Sleeve Gastrectomy in Obese Chinese Women. Obes. Surg. 2019, 29, 2862–2868. [Google Scholar] [CrossRef] [PubMed]
  197. Xu, Y.; Ma, X.; Pan, X.; He, X.; Xiao, Y.; Bao, Y. Correlations between serum concentration of three bone-derived factors and obesity and visceral fat accumulation in a cohort of middle aged men and women. Cardiovasc. Diabetol. 2018, 17, 143. [Google Scholar] [CrossRef] [PubMed]
  198. Lim, W.H.; Wong, G.; Lim, E.M.; Byrnes, E.; Zhu, K.; Devine, A.; Pavlos, N.J.; Prince, R.L.; Lewis, J.R. Circulating Lipocalin 2 Levels Predict Fracture-Related Hospitalizations in Elderly Women: A Prospective Cohort Study. J. Bone Miner. Res. 2015, 30, 2078–2085. [Google Scholar] [CrossRef]
  199. Kim, H.-J.; Ohk, B.; Kang, W.Y.; Seong, S.J.; Suk, K.; Lim, M.-S.; Kim, S.-Y.; Yoon, Y.-R. Deficiency of Lipocalin-2 Promotes Proliferation and Differentiation of Osteoclast Precursors via Regulation of c-Fms Expression and Nuclear Factor-kappa B Activation. J. Bone Metab. 2016, 23, 8–15. [Google Scholar] [CrossRef]
  200. Villalvilla, A.; García-Martín, A.; Largo, R.; Gualillo, O.; Herrero-Beaumont, G.; Gómez, R. The adipokine lipocalin-2 in the context of the osteoarthritic osteochondral junction. Sci. Rep. 2016, 6, 29243. [Google Scholar] [CrossRef]
  201. Costa, D.; Lazzarini, E.; Canciani, B.; Giuliani, A.; Spanò, R.; Marozzi, K.; Manescu, A.; Cancedda, R.; Tavella, S. Altered bone development and turnover in transgenic mice over-expressing Lipocalin-2 in bone. J. Cell. Physiol. 2013, 228, 2210–2221. [Google Scholar] [CrossRef]
  202. Rucci, N.; Capulli, M.; Piperni, S.G.; Cappariello, A.; Lau, P.; Frings-Meuthen, P.; Heer, M.; Teti, A. Lipocalin 2: A New Mechanoresponding Gene Regulating Bone Homeostasis. J. Bone Miner. Res. 2015, 30, 357–368. [Google Scholar] [CrossRef]
  203. Pisoschi, A.M.; Pop, A. The role of antioxidants in the chemistry of oxidative stress: A review. Eur. J. Med. Chem. 2015, 97, 55–74. [Google Scholar] [CrossRef]
  204. Murdolo, G.; Piroddi, M.; Luchetti, F.; Tortoioli, C.; Canonico, B.; Zerbinati, C.; Galli, F.; Iuliano, L. Oxidative stress and lipid peroxidation by-products at the crossroad between adipose organ dysregulation and obesity-linked insulin resistance. Biochimie 2013, 95, 585–594. [Google Scholar] [CrossRef] [PubMed]
  205. Manna, P.; Jain, S.K. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef] [PubMed]
  206. Engin, A. The pathogenesis of obesity-associated adipose tissue inflammation. In Obesity and Lipotoxicity; Springer: Berlin/Heidelberg, Germany, 2017; pp. 221–245. [Google Scholar]
  207. Lee, Y.S.; Kim, A.Y.; Choi, J.W.; Kim, M.; Yasue, S.; Son, H.J.; Masuzaki, H.; Park, K.S.; Kim, J.B. Dysregulation of Adipose Glutathione Peroxidase 3 in Obesity Contributes to Local and Systemic Oxidative Stress. Mol. Endocrinol. 2008, 22, 2176–2189. [Google Scholar] [CrossRef] [PubMed]
  208. Chattopadhyay, M.; Khemka, V.K.; Chatterjee, G.; Ganguly, A.; Mukhopadhyay, S.; Chakrabarti, S. Enhanced ROS production and oxidative damage in subcutaneous white adipose tissue mitochondria in obese and type 2 diabetes subjects. Mol. Cell. Biochem. 2014, 399, 95–103. [Google Scholar] [CrossRef]
  209. Yang, Y.; Sun, Y.; Mao, W.-W.; Zhang, H.; Ni, B.; Jiang, L. Oxidative stress induces downregulation of TP53INP2 and suppresses osteogenic differentiation of BMSCs during osteoporosis through the autophagy degradation pathway. Free. Radic. Biol. Med. 2021, 166, 226–237. [Google Scholar] [CrossRef]
  210. Kanda, Y.; Hinata, T.; Kang, S.W.; Watanabe, Y. Reactive oxygen species mediate adipocyte differentiation in mesenchymal stem cells. Life Sci. 2011, 89, 250–258. [Google Scholar] [CrossRef]
  211. Dong, X.; Bi, L.; He, S.; Meng, G.; Wei, B.; Jia, S.; Liu, J. FFAs-ROS-ERK/P38 pathway plays a key role in adipocyte lipotoxicity on osteoblasts in co-culture. Biochimie 2014, 101, 123–131. [Google Scholar] [CrossRef]
  212. Mukherjee, R.; Moreno-Fernandez, M.E.; Giles, D.A.; Cappelletti, M.; Stankiewicz, T.E.; Chan, C.C.; Divanovic, S. Nicotinamide adenine dinucleotide phosphate (reduced) oxidase 2 modulates inflammatory vigor during nonalcoholic fatty liver disease progression in mice. Hepatol. Commun. 2018, 2, 546–560. [Google Scholar] [CrossRef]
  213. Rahman, M.; El Jamali, A.; Halade, G.V.; Ouhtit, A.; Abou-Saleh, H.; Pintus, G. Nox2 Activity Is Required in Obesity-Mediated Alteration of Bone Remodeling. Oxidative Med. Cell. Longev. 2018, 2018, 1–10. [Google Scholar] [CrossRef]
  214. Kang, I.S.; Kim, C. NADPH oxidase gp91phox contributes to RANKL-induced osteoclast differentiation by upregulating NFATc1. Sci. Rep. 2016, 6, 38014. [Google Scholar] [CrossRef]
  215. Shen, C.-L.; Cao, J.J.; Dagda, R.Y.; Chanjaplammootil, S.; Lu, C.; Chyu, M.-C.; Gao, W.; Wang, J.-S.; Yeh, J.K. Green tea polyphenols benefits body composition and improves bone quality in long-term high-fat diet–induced obese rats. Nutr. Res. 2012, 32, 448–457. [Google Scholar] [CrossRef] [PubMed]
  216. Cao, J.J.; Picklo, M.J. N-Acetylcysteine Supplementation Decreases Osteoclast Differentiation and Increases Bone Mass in Mice Fed a High-Fat Diet. J. Nutr. 2013, 144, 289–296. [Google Scholar] [CrossRef] [PubMed]
  217. Hu, X.; Li, B.; Wu, F.; Liu, X.; Liu, M.; Wang, C.; Shi, Y.; Ye, L. GPX7 Facilitates BMSCs Osteoblastogenesis via ER Stress and mTOR Pathway. J. Cell. Mol. Med. 2021, 25, 10454–10465. [Google Scholar] [CrossRef] [PubMed]
  218. Chang, Y.-C.; Yu, Y.-H.; Shew, J.-Y.; Lee, W.-J.; Hwang, J.-J.; Chen, Y.-H.; Chen, Y.-R.; Wei, P.-C.; Chuang, L.-M.; Lee, W.-H. Deficiency of NPGP x, an oxidative stress sensor, leads to obesity in mice and human. EMBO Mol. Med. 2013, 5, 1165–1179. [Google Scholar] [CrossRef]
  219. Matoušková, P.; Hanousková, B.; Skálová, L. MicroRNAs as Potential Regulators of Glutathione Peroxidases Expression and Their Role in Obesity and Related Pathologies. Int. J. Mol. Sci. 2018, 19, 1199. [Google Scholar] [CrossRef]
  220. He, H.; Zhang, Y.; Sun, Y.; Zhang, Y.; Xu, J.; Yang, Y.; Chen, J. Folic Acid Attenuates High-Fat Diet-Induced Osteoporosis Through the AMPK Signaling Pathway. Front. Cell Dev. Biol. 2022, 9. [Google Scholar] [CrossRef]
  221. Narimiya, T.; Kanzaki, H.; Yamaguchi, Y.; Wada, S.; Katsumata, Y.; Tanaka, K.; Tomonari, H. Nrf2 activation in osteoblasts suppresses osteoclastogenesis via inhibiting IL-6 expression. Bone Rep. 2019, 11, 100228. [Google Scholar] [CrossRef]
  222. Tang, X.; Ma, S.; Li, Y.; Sun, Y.; Zhang, K.; Zhou, Q.; Yu, R. Evaluating the Activity of Sodium Butyrate to Prevent Osteoporosis in Rats by Promoting Osteal GSK-3β/Nrf2 Signaling and Mitochondrial Function. J. Agric. Food Chem. 2020, 68, 6588–6603. [Google Scholar] [CrossRef]
  223. Chan, J.J.; Tay, Y. Noncoding RNA:RNA Regulatory Networks in Cancer. Int. J. Mol. Sci. 2018, 19, 1310. [Google Scholar] [CrossRef]
  224. Dykes, I.M.; Emanueli, C. Transcriptional and Post-transcriptional Gene Regulation by Long Non-coding RNA. Genom. Proteom. Bioinform. 2017, 15, 177–186. [Google Scholar] [CrossRef]
  225. Wang, L.; Cho, K.B.; Li, Y.; Tao, G.; Xie, Z.; Guo, B. Long Noncoding RNA (lncRNA)-Mediated Competing Endogenous RNA Networks Provide Novel Potential Biomarkers and Therapeutic Targets for Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 5758. [Google Scholar] [CrossRef] [Green Version]
  226. Wijesinghe, S.N.; Nicholson, T.; Tsintzas, K.; Jones, S.W. Involvements of long noncoding RNAs in obesity-associated inflammatory diseases. Obes. Rev. 2020, 22. [Google Scholar] [CrossRef] [PubMed]
  227. Mugoni, V.; Ciani, Y.; Nardella, C.; Demichelis, F. Circulating RNAs in prostate cancer patients. Cancer Lett. 2021, 524, 57–69. [Google Scholar] [CrossRef] [PubMed]
  228. Xu, F.; Jin, L.; Jin, Y.; Nie, Z.; Zheng, H. Long noncoding RNAs in autoimmune diseases. J. Biomed. Mater. Res. Part A 2018, 107, 468–475. [Google Scholar] [CrossRef] [PubMed]
  229. Gurha, P. Noncoding RNAs in cardiovascular diseases. Curr. Opin. Cardiol. 2019, 34, 241–245. [Google Scholar] [CrossRef] [PubMed]
  230. Yu, L.; Xia, K.; Cen, X.; Huang, X.; Sun, W.; Zhao, Z.; Liu, J. DNA methylation of noncoding RNAs: New insights into osteogenesis and common bone diseases. Stem Cell Res. Ther. 2020, 11, 109. [Google Scholar] [CrossRef]
  231. Endisha, H.; Datta, P.; Sharma, A.; Nakamura, S.; Rossomacha, E.; Younan, C.; Ali, S.A.; Tavallaee, G.; Lively, S.; Potla, P.; et al. MicroRNA-34a-5p Promotes Joint Destruction During Osteoarthritis. Arthritis Rheumatol. 2020, 73, 426–439. [Google Scholar] [CrossRef]
  232. Pan, Y.; Hui, X.; Hoo, R.L.C.; Ye, D.; Chan, C.Y.C.; Feng, T.; Wang, Y.; Lam, K.S.L.; Xu, A. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J. Clin. Investig. 2019, 129, 834–849. [Google Scholar] [CrossRef]
  233. Yuan, T.; Shi, C.; Xu, W.; Yang, H.-L.; Xia, B.; Tian, C. Extracellular vesicles derived from T-cell acute lymphoblastic leukemia inhibit osteogenic differentiation of bone marrow mesenchymal stem cells via miR-34a-5p. Endocr. J. 2021, 68, 1197–1208. [Google Scholar] [CrossRef]
  234. Nóbrega, O.T.; Morais-Junior, G.S.; Viana, N.I.; Reis, S.T.; Perez, D.I.V.; Freitas, W.M.; Sposito, A.C.; Leite, K.R.M.; Srougi, M. Circulating miR-34a and Bone Mineral Density of Brazilian Very-Old Adults. J. Aging Res. 2020, 2020, 3431828. [Google Scholar] [CrossRef]
  235. Gu, Y.; Ma, L.; Song, L.; Li, X.; Chen, D.; Bai, X. miR-155 Inhibits Mouse Osteoblast Differentiation by Suppressing SMAD5 Expression. BioMed Res. Int. 2017, 2017, 1893520. [Google Scholar] [CrossRef] [Green Version]
  236. Wu, T.; Xie, M.; Wang, X.; Jiang, X.; Li, J.; Huang, H. miR-155 modulates TNF-α-inhibited osteogenic differentiation by targeting SOCS1 expression. Bone 2012, 51, 498–505. [Google Scholar] [CrossRef] [PubMed]
  237. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384.e12. [Google Scholar] [CrossRef]
  238. Zhang, Y.; Mei, H.; Chang, X.; Chen, F.; Zhu, Y.; Han, X. Adipocyte-derived microvesicles from obese mice induce M1 macrophage phenotype through secreted miR-155. J. Mol. Cell Biol. 2016, 8, 505–517. [Google Scholar] [CrossRef] [PubMed]
  239. Sun, Y.; Xu, J.; Xu, L.; Zhang, J.; Chan, K.; Pan, X.; Li, G. MiR-503 Promotes Bone Formation in Distraction Osteogenesis through Suppressing Smurf1 Expression. Sci. Rep. 2017, 7, 409. [Google Scholar] [CrossRef] [PubMed]
  240. Man, X.-F.; Tan, S.-W.; Tang, H.-N.; Guo, Y.; Tang, C.-Y.; Tang, J.; Zhou, C.-L.; Zhou, H.-D. MiR-503 inhibits adipogenesis by targeting bone morphogenetic protein receptor 1a. Am. J. Transl. Res. 2016, 8, 2727–2737. [Google Scholar]
  241. Yue, H.-Q.; Zhou, Y.-H.; Guo, Y.; Tang, C.-Y.; Wang, F.; Zhou, H.-D. Serum miR-503 is a Candidate Biomarker for Differentiating Metabolic Healthy Obesity from Metabolic Unhealthy Obesity. Diabetes Metab. Syndr. Obesity Targets Ther. 2020, 13, 2667–2676. [Google Scholar] [CrossRef]
  242. Li, Y.; Yang, F.; Gao, M.; Gong, R.; Jin, M.; Liu, T.; Sun, Y.; Fu, Y.; Huang, Q.; Zhang, W.; et al. miR-149-3p Regulates the Switch between Adipogenic and Osteogenic Differentiation of BMSCs by Targeting FTO. Mol. Ther.-Nucleic Acids 2019, 17, 590–600. [Google Scholar] [CrossRef]
  243. Chartoumpekis, D.V.; Zaravinos, A.; Ziros, P.G.; Iskrenova, R.P.; Psyrogiannis, A.I.; Kyriazopoulou, V.E.; Habeos, I.G. Differential Expression of MicroRNAs in Adipose Tissue after Long-Term High-Fat Diet-Induced Obesity in Mice. PLoS ONE 2012, 7, e34872. [Google Scholar] [CrossRef]
  244. Lou, Z.; Peng, Z.; Wang, B.; Li, X.; Li, X.; Zhang, X. miR-142-5p promotes the osteoclast differentiation of bone marrow-derived macrophages via PTEN/PI3K/AKT/FoxO1 pathway. J. Bone Miner. Metab. 2019, 37, 815–824. [Google Scholar] [CrossRef]
  245. Teng, Z.; Xie, X.; Zhu, Y.; Liu, J.; Hu, X.; Na, Q.; Zhang, X.; Wei, G.; Xu, S.; Liu, Y.; et al. miR-142-5p in Bone Marrow-Derived Mesenchymal Stem Cells Promotes Osteoporosis Involving Targeting Adhesion Molecule VCAM-1 and Inhibiting Cell Migration. BioMed Res. Int. 2018, 2018, 3274641. [Google Scholar] [CrossRef] [PubMed]
  246. Mao, Z.; Zhu, Y.; Hao, W.; Chu, C.; Su, H. MicroRNA-155 inhibition up-regulates LEPR to inhibit osteoclast activation and bone resorption via activation of AMPK in alendronate-treated osteoporotic mice. IUBMB Life 2019, 71, 1916–1928. [Google Scholar] [CrossRef] [PubMed]
  247. Zhang, J.; Zhao, H.; Chen, J.; Xia, B.; Jin, Y.; Wei, W.; Shen, J.; Huang, Y. Interferon-β-induced miR-155 inhibits osteoclast differentiation by targeting SOCS1 and MITF. FEBS Lett. 2012, 586, 3255–3262. [Google Scholar] [CrossRef]
  248. Wang, Y.; Chen, W.; Zhao, L.; Li, Y.; Liu, Z.; Gao, H.; Bai, X.; Wang, B. Obesity regulates miR-467/HoxA10 axis on osteogenic differentiation and fracture healing by BMSC-derived exosome LncRNA H19. J. Cell. Mol. Med. 2021, 25, 1712–1724. [Google Scholar] [CrossRef] [PubMed]
  249. Zhao, X.; Yang, Y.; Sun, B.F.; Zhao, Y.L.; Yang, Y.G. FTO and obesity: Mechanisms of association. Curr. Diabetes Rep. 2014, 14, 486. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Changes in adipose tissue during obesity. In lean subjects, adipose tissue presents an anti-inflammatory state. Moreover, adipocytes present average triglyceride storage. During obesity, nutrient excess and low expenditure promote a hypertrophic state in adipocytes. The hypertrophic adipocytes and pro-inflammatory immune cells, such as M1-like macrophages, neutrophils, and CD8+ T cells, contribute to a low-grade inflammatory state in subjects with obesity. Most of the pro-inflammatory factors released by obese adipose tissue interfere with bone cells leading to bone loss.
Figure 1. Changes in adipose tissue during obesity. In lean subjects, adipose tissue presents an anti-inflammatory state. Moreover, adipocytes present average triglyceride storage. During obesity, nutrient excess and low expenditure promote a hypertrophic state in adipocytes. The hypertrophic adipocytes and pro-inflammatory immune cells, such as M1-like macrophages, neutrophils, and CD8+ T cells, contribute to a low-grade inflammatory state in subjects with obesity. Most of the pro-inflammatory factors released by obese adipose tissue interfere with bone cells leading to bone loss.
Cells 12 00521 g001
Figure 2. Obese adipose tissue-derived proinflammatory cytokines on bone remodeling. Pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, released by obese adipose tissue, exert effects on osteoclast precursors, increasing osteoclast differentiation and resorption. Moreover, these cytokines reduce the osteogenic differentiation of mesenchymal stem cells and compromise osteoblast bone formation. TNF-α, IL-6, and IL-1β also promote apoptosis and increased expression of osteoclast stimulation factors by osteocytes. The unbalanced activities between osteoblasts and osteoclasts induce osteoporosis.
Figure 2. Obese adipose tissue-derived proinflammatory cytokines on bone remodeling. Pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, released by obese adipose tissue, exert effects on osteoclast precursors, increasing osteoclast differentiation and resorption. Moreover, these cytokines reduce the osteogenic differentiation of mesenchymal stem cells and compromise osteoblast bone formation. TNF-α, IL-6, and IL-1β also promote apoptosis and increased expression of osteoclast stimulation factors by osteocytes. The unbalanced activities between osteoblasts and osteoclasts induce osteoporosis.
Cells 12 00521 g002
Figure 3. Impacts of gut alterations and dysbiosis on bone remodeling. Obesity promotes an increased permeability of the intestinal barrier, which leads to enhanced serum LPS concentrations. High endotoxin levels and the altered gut microbiota, with increased Firmicutes/Bacteroidetes ratio and decreased frequency of beneficial microorganisms, contribute to increased bone resorption and decreased bone formation in obesity.
Figure 3. Impacts of gut alterations and dysbiosis on bone remodeling. Obesity promotes an increased permeability of the intestinal barrier, which leads to enhanced serum LPS concentrations. High endotoxin levels and the altered gut microbiota, with increased Firmicutes/Bacteroidetes ratio and decreased frequency of beneficial microorganisms, contribute to increased bone resorption and decreased bone formation in obesity.
Cells 12 00521 g003
Figure 4. Obesity’s effects on osteokines secretion and bone loss. Obesity promotes bone dysfunctions that alter osteokine secretion. Serum uncarboxylated osteocalcin is reduced in subjects with obesity, while FGF-23 concentration is enhanced. Thus, this pathologic condition aggravates insulin resistance and weight gain. The worsening of obesity leads to more severe bone damage, becoming a vicious cycle. Lipocalin-2, which is also produced by bone cells, may exert anti-obesity effects, but the participation of lipocalin 2 in obesity-associated bone loss still needs to be fully understood.
Figure 4. Obesity’s effects on osteokines secretion and bone loss. Obesity promotes bone dysfunctions that alter osteokine secretion. Serum uncarboxylated osteocalcin is reduced in subjects with obesity, while FGF-23 concentration is enhanced. Thus, this pathologic condition aggravates insulin resistance and weight gain. The worsening of obesity leads to more severe bone damage, becoming a vicious cycle. Lipocalin-2, which is also produced by bone cells, may exert anti-obesity effects, but the participation of lipocalin 2 in obesity-associated bone loss still needs to be fully understood.
Cells 12 00521 g004
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

Forte, Y.S.; Renovato-Martins, M.; Barja-Fidalgo, C. Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells 2023, 12, 521. https://doi.org/10.3390/cells12040521

AMA Style

Forte YS, Renovato-Martins M, Barja-Fidalgo C. Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells. 2023; 12(4):521. https://doi.org/10.3390/cells12040521

Chicago/Turabian Style

Forte, Yasmin Silva, Mariana Renovato-Martins, and Christina Barja-Fidalgo. 2023. "Cellular and Molecular Mechanisms Associating Obesity to Bone Loss" Cells 12, no. 4: 521. https://doi.org/10.3390/cells12040521

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