International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Osteoporosis, a prevalent metabolic bone disease with notable medical and socioeconomic implications, is characterized by low bone mass and microarchitectural deterioration, leading to weakened bone strength and an increased risk of fragility fractures (1). According to the National Health and Nutrition Examination Survey in the United States, ~20% of postmenopausal women have been diagnosed with osteoporosis (2). As the global population ages, this prevalence is expected to increase in the coming years (3). For individuals with osteoporosis, the lifetime risk of fractures can reach up to 40%; from the perspective of patients, fractures, along with the subsequent loss of mobility and independence, often result in a substantial decline in quality of life. Moreover, osteoporotic fractures, particularly of the hip and spine, are associated with a 12-month excess mortality rate of up to 20%, primarily due to hospitalization, which increases the risk of complications such as pneumonia or thromboembolic events from prolonged immobility, further compounding the public health burden (4). The pathophysiology of osteoporosis is marked by an imbalance between bone resorption and formation, which is influenced by factors such as aging, hormonal changes, nutritional deficiencies and genetics (5,6). Notably, the interaction between osteoblasts and osteoclasts is crucial for maintaining bone homeostasis. Osteoblasts express and secrete receptor activator of NF-κB ligand (RANKL), which promotes the differentiation of osteoclast precursors into mature osteoclasts. Conversely, when bone resorption disrupts the bone structure, osteoclasts release insulin-like growth factors (IGFs), which stimulate osteogenic differentiation (7). However, dysregulated bone remodeling negatively affects bone mass. A reduction in osteoblast activity, responsible for producing the calcified organic extracellular matrix, leads to increased bone resorption. Simultaneously, osteoclasts degrade the extracellular matrix and the cumulative effect of these processes can result in bone loss (8). Understanding the underlying mechanisms of bone remodeling and the factors influencing bone mass is critical for developing effective strategies to prevent and treat osteoporosis.
Branched-chain amino acids (BCAAs), including leucine, isoleucine and valine, are essential amino acids primarily known for their role in muscle protein synthesis (9). In addition to their function in skeletal muscle, BCAAs have been linked to the regulation of metabolic disorders such as diabetes, cardiovascular diseases and cancer (10). They may also influence aging and longevity in animal models (11). Recent research has identified a range of molecules involved in maintaining bone homeostasis, with BCAAs emerging as potential modulators within this regulatory network (12).
The present review summarizes the interactions between bone metabolism and BCAA metabolism, focusing on the causes and consequences of metabolic abnormalities. It also explores the relationship between bone metabolism and osteoporosis, highlighting how dysregulated BCAA metabolism may contribute to osteoporosis. Finally, the potential of BCAAs as a treatment for osteoporosis is discussed, offering novel insights into their role in bone health. In conclusion, the current review may enhance understanding of the mechanisms by which BCAAs contribute to the pathogenesis of osteoporosis. It not only provides dietary recommendations but also emphasizes the potential for BCAA-based interventions in the management of osteoporosis, paving the way for novel therapeutic approaches to combat this disease.
Bone is a dynamic, metabolically active tissue and an essential organ system in higher vertebrates, serving as the primary structural component of the skeleton. It is a specialized connective tissue composed of osteocytes, osteoblasts, bone-lining cells and osteoclasts. The bone matrix consists of ~60% inorganic mineral phase (primarily semi-crystalline, carbonated hydroxyapatite), 30% organic phase, which includes type I collagen fibrils and non-collagenous proteins, and 10% water (13). Additionally, small leucine-rich proteoglycans, a key component of non-collagenous proteins, are present in the bone matrix (14).
Bone performs four critical functions: i) Providing structural support and anchorage for muscles, ii) protecting vital organs such as the brain and bone marrow, iii) supporting hematopoiesis (15-17), and iv) serving as a metabolic reservoir for calcium and phosphate. These physiological functions rely on the tightly regulated processes of bone modeling and remodeling to maintain skeletal homeostasis (18).
Bone remodeling involves the resorption of old or damaged bone by osteoclasts, followed by the formation of new bone by osteoblasts (19). In contrast to bone modeling, remodeling is a coupled, spatially and temporally coordinated process that does not alter bone shape (20). It is a tightly regulated physiological process and dysfunction in any component of this system can disrupt bone metabolism, contributing to conditions such as osteoporosis (21).
Bone metabolism is a complex process precisely coordinated by osteoclasts, osteoblasts and osteocytes. Osteoclasts originate from mononuclear precursors of the monocyte/macrophage lineage, and their differentiation and activation depend on two critical signals: Macrophage colony-stimulating factor (22) and RANKL (23,24), as substantiated by Nakahama et al (25). The binding of RANKL to receptor activator of NF-κB (RANK) on osteoclast precursors initiates osteoclastogenesis (26). Mature, functional osteoclasts form a ruffled border mediated by Src kinase (27,28) and secrete cathepsin K to degrade the bone matrix, accomplishing bone resorption (29,30). Gelb et al (29) demonstrated that osteoclasts from human patients with cathepsin K gene mutations fail to efficiently degrade the bone organic matrix, providing direct clinical evidence linking cathepsin K to osteoclastic resorption.
Osteoblasts are derived from various sources, including bone marrow stromal cells and bone-lining cells (31). During bone resorption, factors released from the bone matrix, such as transforming growth factor-β (TGF-β) and IGF-1, recruit osteoblast precursors to the bone surface (32). Osteoblast differentiation is primarily regulated by the canonical Wnt/β-catenin signaling pathway. Upon activation of this pathway, β-catenin is stabilized and translocates into the nucleus, where it binds to LEF/TCF transcription factors. This interaction activates the expression of osteogenic genes such as Runx2, promoting the differentiation of osteoblasts from progenitor cells into mature cells (33). Mature osteoblasts then secrete type I collagen and other components, forming osteoid and facilitating its mineralization to build new bone. After completing their task, most osteoblasts undergo apoptosis, while some differentiate into osteocytes or revert to quiescent bone-lining cells (34,35).
Osteocytes, embedded within the mineralized matrix, serve as key regulators of bone homeostasis (36). By contrast, necrotic osteocytes release damage-associated molecular patterns (37), which bind to pattern recognition receptors (38), initiating the production of proinflammatory cytokines, such as TNFα, IL-1 and IL-6 (39). These cytokines indirectly amplify RANKL signaling (40) by increasing RANKL expression in osteoblasts (41). Osteocyte apoptosis is a critical trigger for bone remodeling (37), as these cells recruit osteoclasts to microcracks, initiating targeted bone resorption and maintaining dynamic bone tissue homeostasis (42). Kringelbach et al (41) demonstrated that apoptosis in the MLO-Y4 osteocyte cell line induces the release of adenosine triphosphate (ATP) via the activation of Pannexin 1 (PANX1) channels. A similar phenomenon has been observed in mice treated with a P2X7 receptor (P2X7R) inhibitor, highlighting the role of P2X7R as a coactivator of PANX1 (43). The released ATP subsequently stimulates RANKL expression through a signaling cascade involving the purinergic receptor P2Y (P2Y)1 receptor and cyclooxygenase activity (44-46). ATP released from apoptotic osteocytes likely binds to P2Y2 receptors on osteocytes in the penumbra regions, known as 'bystander' osteocytes (47); this binding may also trigger RANKL production and release (43).
The termination of bone remodeling involves multiple steps, including the secretion of osteoprotegerin (OPG) by mature osteoblasts and/or osteocytes, which may serve as a key inhibitory signal (48). Additionally, upregulation of the anti-apoptotic protein BCL-2 in 'bystander' osteocytes near the damage site helps protect viable osteocytes from osteoclastogenic stimuli originating from adjacent apoptotic cells (Fig. 1) (49,50).
Bone metabolism is regulated by both local intercellular signaling among bone cells and a range of systemic factors. Calcium and phosphate regulators maintain mineral homeostasis, which is crucial for bone mineralization. 1,25(OH)2D3 enhances intestinal calcium and phosphate absorption directly (51), while also modulating osteoblast and osteoclast activity (52-54). Concurrently, parathyroid hormone (PTH) and PTH-related peptide regulate calcium-phosphate homeostasis in bone and the kidneys by activating the cyclic adenosine monophosphate-protein kinase A pathway through the PTH1 receptor (55,56). Elevated phosphate levels, in turn, stimulate the secretion of fibroblast growth factor 23, which suppresses 1,25(OH)2D3 synthesis and promotes renal phosphate excretion, creating a critical feedback loop (57,58). Calcitonin acts antagonistically by directly inhibiting bone resorption to lower blood calcium levels (59,60), thereby ensuring precise regulation of calcium homeostasis.
Osteocyte communication and the integrity of the network are central to the mechanosensing and signaling system of bone. Connexin 43 (Cx43) underpins intercellular communication among osteocytes, and its loss of function directly impairs bone mass acquisition and maintenance (61-63). Similarly, the integrity of the osteocyte dendritic network is essential, with age-related declines in network function directly associated with cortical bone deterioration (64). Additionally, TGF-β1 signaling is a key intrinsic regulator of this network, preserving bone mechanical integrity by modulating dendritic length and the expression of genes such as sclerostin and MMPs (65). These factors, particularly in aging and metabolic diseases (including diabetes and obesity), interact with the broader bone metabolism regulatory network (66).
The balance between bone formation and resorption directly determines the net outcome of bone remodeling. For example, growth hormone, by stimulating IGF-1 expression, increases bone turnover with a net anabolic effect, leading to modest bone mass gain (67,68). Estrogen promotes osteocyte survival via semaphorin 3A (69), fluid flow shear stress (70), Wnt/β-catenin signaling (71) and Cx43 expression (72), helping to counteract age-related bone loss. The myokine irisin also serves a protective role in the bone-muscle axis by reducing osteocyte apoptosis (73). Additionally, vitamin K-dependent osteocalcin, secreted by osteoblasts, connects bone metabolism to systemic energy metabolism, broadening the regulation of bone homeostasis (Fig. 2) (74).
Bone metabolic imbalance is a complex condition arising from a variety of physiological and pathological factors. A primary underlying mechanism is the disruption of the dynamic balance between osteoclastic bone resorption and osteoblastic bone formation. This dysregulation can lead to skeletal disorders such as osteoporosis, which is characterized by reduced bone mass and increased susceptibility to fractures. Contributing factors include hormonal imbalances, inadequate nutrition and genetic predispositions (75).
Hormonal changes, particularly those involving estrogen and PTH, serve a central role in bone metabolism. Estrogen deficiency, commonly observed in postmenopausal women, accelerates bone resorption, thereby promoting osteoporosis (76). Similarly, parathyroid gland disorders disrupt calcium and phosphate metabolism, further exacerbating bone loss (77). The skeleton also functions as an endocrine organ; hormones such as osteocalcin regulate both energy metabolism and bone homeostasis, highlighting the close link between skeletal and metabolic health (78).
Nutritional factors markedly influence bone health. Adequate calcium and vitamin D intake is essential for maintaining bone density, with deficiencies in these nutrients leading to impaired bone mineralization and an increased fracture risk (79). Additionally, while high protein intake exerts a bone-anabolic effect, this benefit can be counteracted by a high dietary acid load, ultimately promoting bone loss (80). The role of vitamin K, particularly in relation to osteocalcin, is also important; vitamin K is involved in the carboxylation of osteocalcin, a process essential for bone mineralization and metabolic regulation (74).
Genetic factors and metabolic disorders predispose individuals to bone metabolism disorders. Conditions such as hypophosphatasia and lysosomal storage disease result in skeletal abnormalities and heightened osteoporosis risk. These genetic disorders often cause imbalances in calcium and phosphate metabolism, which are critical for bone health (81). Additionally, abnormalities in purine metabolism have been identified as a notable factor in bone remodeling, contributing to osteoporosis in various high-risk populations (82).
Environmental and lifestyle factors, such as physical inactivity, smoking and excessive alcohol consumption, are well-established negative influences on bone health. These factors increase oxidative stress and inflammation, both of which are detrimental to bone remodeling (83-85). The accumulation of advanced oxidation protein products has been shown to contribute to bone-fat imbalance during skeletal aging, highlighting the impact of oxidative stress on bone metabolism (Table I) (86-88).
BCAAs, comprising leucine, isoleucine and valine, are essential amino acids that must be sourced from the diet, as they cannot be synthesized endogenously in animals (89). Despite being classified together as BCAAs, leucine, isoleucine and valine exhibit distinct structural, metabolic and functional characteristics, which underlie their intricate regulation of bone metabolism (90). Leucine, with its highly hydrophobic isobutyl side chain, is the most hydrophobic of the three, a property essential for its function as a signaling molecule. This hydrophobicity facilitates the role of leucine in activating downstream targets, establishing its dominant role in signal transduction (91).
Beyond serving as fundamental components for protein synthesis, BCAAs are key regulators of cellular metabolism, energy production and signal transduction (92). After ingestion, BCAAs undergo enzymatic transformation across various tissues, with the highest concentrations observed in skeletal muscle, brown adipose tissue, liver, kidney and heart (93). Enzymes such as BCAA transaminase (BCAT) and branched-chain α-ketoacid dehydrogenase (BCKDH) catalyze the production of important metabolites, including branched-chain α-ketoacids (BCKAs), acetyl-CoA and succinyl-CoA, which contribute to processes such as the tricarboxylic acid cycle and fatty acid metabolism (Fig. 3) (94). The distinct metabolic pathways of the three BCAAs suggest that they may exert differential effects on bone metabolism by regulating cellular energy status, metabolite pools and signal transduction pathways.
As critical nutrient signals, BCAAs modulate cellular metabolism and energy production. They activate anabolic pathways, particularly the mechanistic target of rapamycin (mTOR) signaling pathway, which promotes protein synthesis in skeletal muscle and enhances immune cell proliferation and function, including T cells and natural killer cells (95). Additionally, emerging evidence has highlighted a 'muscle-bone axis', where skeletal muscle and bone tissue interact bidirectionally (96). Immune cells, including osteoclast precursors and macrophages, also serve pivotal roles in bone resorption and remodeling (97). By influencing both the musculoskeletal and immune systems, BCAAs likely exert indirect yet notable effects on bone metabolism and homeostasis. Further research is needed to elucidate the direct regulatory roles of BCAAs in bone metabolism, particularly through pathways involving cellular metabolism and energy balance.
BCAAs and their metabolic derivatives function as molecular signals in bone metabolism. Both leucine (98) and valine (99) activate the mTOR complex 1 (mTORC1) pathway. The biochemical pathways of BCAA metabolism are well-characterized, and preclinical studies have indicated that inhibition of mTOR suppresses osteoclast differentiation and survival, potentially impairing bone resorption (100-103). These findings are further supported by the BOLERO-2 trial, where everolimus, a clinically used mTOR inhibitor, demonstrated beneficial effects on bone in patients with advanced breast cancer (104). Metabolic derivatives of valine, such as β-aminoisobutyric acid (BAIBA) and 3-hydroxyisobutyric acid (3-HIB), act as myo-osseous metabolic messengers, regulating bone homeostasis through distinct signaling pathways. BAIBA, regulated by peroxisome proliferator-activated receptor (PPAR)γ coactivator-1α (PGC-1α), activates the PPARα pathway, inducing white adipose tissue browning, improving insulin sensitivity and potentially enhancing osteogenesis while inhibiting osteoclastogenesis (105). Conversely, 3-HIB promotes trans-endothelial fatty acid transport via FATP3/4, leading to lipotoxicity and insulin resistance, which may disrupt bone homeostasis (106). Further research is needed to better define the direct roles of BCAAs in bone metabolism through these signal transduction pathways, supported by additional well-designed clinical studies.
Metabolomic analyses from large randomized controlled trials have established circulating BCAA levels as key indicators of metabolic health. These levels, beyond body mass index, predict the risk of insulin resistance, cardiovascular diseases and diabetes, and reveal ethnic differences in obesity-associated metabolic disturbances (107,108). A Mendelian randomization study further indicated that elevated BCAA levels are linked to an increased risk of type 2 diabetes (109). Collectively, these findings suggest that dysregulated BCAA metabolism contributes to the pathogenesis of insulin resistance, obesity and diabetes (110,111). Moreover, insulin resistance may promote BCAA accumulation (112,113), creating a self-perpetuating cycle. Insulin resistance, frequently accompanied by altered BCAA metabolism, adversely affects bone health by impairing osteoblast function and inhibiting bone formation (Fig. 3) (114).
Disorders in BCAA metabolism contribute to a complex pathological network that impacts bone metabolism. This network involves both direct regulation of bone cells (osteoblasts and osteoclasts) (115), and indirect effects mediated by systemic metabolic disturbances, ultimately disrupting bone homeostasis (114,116).
A key mechanism through which BCAA metabolism directly influences bone metabolism is its role in osteoclast maturation. Studies have shown that inhibition of BCAT1, a key enzyme in BCAA metabolism, effectively reduces osteoclast maturation (12,117). BCAT1 catalyzes the conversion of BCAAs to their metabolites (BCKAs), which fuel cellular energy production (90) and sustain the expression of essential osteoclastogenic factors (such as NFATc1 and ATP6v0d2) (118), as well as the regulation of fusion-related genes (including DCSTAMP and CD9) (119,120). These processes collectively underlie its critical role in the later stages of osteoclast differentiation. In vivo animal experiments have further confirmed that BCAT1 is central to the osteoclast multinucleation regulatory network. Its deletion markedly inhibits osteoclast activity, increases bone mass and improves bone strength. These results establish a strong link between abnormal BCAA metabolism and bone metabolic disorders mediated by osteoclasts (117).
In contrast to osteoclasts, the impact of BCAA metabolism on osteoblasts is more complex, highlighting the adverse effects of abnormal BCAA metabolism on bone health. Singha et al (121) demonstrated that rapamycin, an mTOR inhibitor, can effectively suppress osteoblast proliferation and differentiation by reducing RUNX2 protein expression, decreasing alkaline phosphatase activity and inhibiting matrix mineralization, supporting the pro-osteogenic role of mTORC1 in osteogenesis. However, dysregulated BCAA metabolism, such as chronic overaccumulation or insufficient breakdown products, may disrupt the mTORC1 pathway (122). Prolonged activation of mTORC1 can induce 'metabolic stress' in osteoblasts, characterized by G1 phase cell cycle arrest and reduced proliferative capacity, ultimately impairing bone metabolism (123). These findings suggest that BCAAs regulate osteoblast differentiation to a certain extent via the mTOR signaling pathway; however, its translational applicability in humans remains to be confirmed through rigorously designed clinical investigations.
High glutamine levels further inhibit the mTORC2/Akt-S473/RUNX2 axis by activating mTORC1, which leads to the ubiquitin-mediated degradation of RUNX2, thereby suppressing osteogenic differentiation (124). These findings indicate that the regulatory role of mTORC1 signaling in osteoblast differentiation may be context-dependent, necessitating additional in vitro experiments or clinical studies to confirm these results.
As aforementioned, dysregulated BCAA metabolism is linked to various metabolic disorders, including obesity and type 2 diabetes, both of which are well-established risk factors for osteoporosis and other bone-related conditions (125). In patients with obesity and type 2 diabetes, elevated BCAA levels are strongly associated with insulin resistance. BCAAs activate the mTORC1 signaling pathway, which decouples insulin signal transduction, thereby exacerbating insulin resistance (114,126). Furthermore, intermediate products of BCAA metabolism, such as BCKAs, inhibit insulin signal transduction, further aggravating metabolic disorders (127). These metabolic disruptions not only affect systemic metabolism but also impair bone homeostasis by disrupting energy metabolism and signal transduction in osteocytes (115,128).
Additionally, BCAAs influence lipid metabolism, and disruptions in this pathway can lead to altered lipid profiles that may negatively affect bone density and strength. Impaired BCAA catabolism has been linked to lipid accumulation, which can adversely impact bone tissue (129,130). BCAA metabolic disorders may worsen the complex pathological network of bone metabolism by modulating oxidative stress and inflammatory responses. Elevated BCAA and fatty acid levels induce mitochondrial dysfunction, increase oxidative stress, and activate inflammatory signaling pathways, all of which can contribute to bone metabolic diseases, such as osteoporosis, by impairing osteocyte function and disrupting the bone microenvironment (131,132). Furthermore, BCAA metabolic disorders may indirectly influence bone metabolism by interfering with lipid metabolism (133).
Regarding the mTORC1 pathway, Lin et al (134) demonstrated that reduced angiopoietin-like-4 levels modulate BCAA metabolism, activating mTOR signaling and promoting accelerated proliferation in osteosarcoma cells. This suggests that disturbances in this pathway, resulting from abnormal BCAA metabolism, may influence bone formation and remodeling, ultimately contributing to bone metabolic disorders.
Osteoporosis is a prevalent metabolic bone disorder characterized by generalized bone mass loss and deterioration of bone microarchitecture, which increases bone fragility and the risk of fragility fractures (135). Notably, osteoporosis also elevates the likelihood of secondary complications, such as pneumonia or thromboembolic events, particularly due to immobilization following a fracture (4). Rather than a singular condition, osteoporosis encompasses a spectrum of distinct pathological states, typically classified as primary or secondary. Primary osteoporosis includes two main subtypes: Type I, or postmenopausal osteoporosis, which predominantly affects women after menopause due to estrogen deficiency, and type II, also known as age-related or senile osteoporosis, which is associated with aging in both sexes. By contrast, secondary osteoporosis arises from various underlying medical conditions, reduced physical activity or the adverse effects of medical treatments (136).
The primary pathological features of osteoporosis include decreased bone mass, disrupted bone microarchitecture and an imbalance in bone remodeling, where bone resorption outpaces bone formation, leading to a net loss of bone tissue (135).
Type I osteoporosis primarily results from estrogen deficiency, which accelerates bone resorption by increasing the RANKL/OPG ratio and enhancing osteoclast activity (137). Several bone metabolism pathways are implicated in this process: i) Estrogen deficiency prolongs the lifespan of bone marrow dendritic cells and increases the secretion of IL-7 and IL-15, which promote the conversion of memory T (TM) cells into effector TM cells. The subsequent release of TNFα and IL-17A acts synergistically with RANKL to stimulate osteoclastogenesis and enhance osteoclast activity. ii) Estrogen deficiency also increases intestinal permeability and triggers inflammation in the intestinal mucosa, resulting in elevated production of T helper 17 (Th17) cells. IL-17A produced by Th17 cells further enhances osteoclast differentiation and bone resorption. Additionally, metabolites from gut microbiota, such as butyric acid, polyamines and other short-chain fatty acids, may indirectly promote osteoclast activity; however, the exact mechanisms remain to be fully elucidated (138).
Vitamin D deficiency impairs calcium absorption, a condition further exacerbated by inadequate dietary calcium intake. This disruption in calcium homeostasis leads to increased bone resorption and heightened bone remodeling. In a large European cohort study of 8,532 postmenopausal women with insufficient vitamin D levels, 97% were found to have osteoporosis (139). Another study revealed that 82% of individuals with osteoporosis had calcium intakes below the recommended daily amount of 1,000 mg (140). Moreover, phosphorus deficiency, a common marker of poor nutritional status in elderly patients, is associated with an elevated risk of fractures (141). Thus, deficiencies in vitamin D, calcium and phosphorus further increase fracture risk in elderly women with osteoporosis.
During the progression of osteoporosis in the elderly, both cellular senescence and epigenetic regulatory pathways serve key roles in modulating bone metabolism. As aging progresses, bone marrow mesenchymal stem cells (BMSCs) undergo senescence, accompanied by an upregulation of nucleosome assembly protein 1 like 2 (NAP1L2) expression. NAP1L2 contributes to bone loss through two primary mechanisms. First, it activates the NF-κB signaling pathway, leading to increased secretion of senescence-associated factors, such as IL-6 and IL-8. These factors exacerbate cellular senescence and impair the osteogenic differentiation of BMSCs, thus negatively impacting bone formation. Second, as a histone chaperone, NAP1L2 recruits sirtuin 1 to deacetylate H3K14ac at the promoters of key osteogenic genes, including Runx2, Sp7 and Bglap, causing their transcriptional repression. This epigenetic modification further inhibits osteogenic differentiation, reduces bone formation, and accelerates the development of osteoporosis (142).
Abnormal cellular functions also contribute to osteoporosis pathogenesis: i) Inhibition of osteoblast differentiation and function, which is associated with downregulation of the BMP/Smad (143) and Wnt/β-catenin (144) pathways, thus preventing MSCs from differentiating into osteoblasts; ii) overactivation of osteoclasts, bone matrix degradation is accelerated due to lysosomal acidification abnormalities, such as increased V-ATPase activity (145) and elevated cathepsin K activity (146); and iii) dysfunction in the osteocyte network, where osteocyte apoptosis and the accumulation of microdamage impair mechanosignal perception, inhibiting bone repair (147). Furthermore, senescence or mechanical stimulation reduces the upregulation of sclerostin, which antagonizes the Wnt pathway and inhibits bone formation (148).
Dysregulated molecular signaling pathways also contribute to osteoporosis, particularly those involved in the RANKL/RANK/OPG system, the Wnt/β-catenin pathway, the BMP-Smad pathway and the PI3K/Akt/mTOR pathway. Additionally, hormonal and cytokine networks, particularly those involving PTH and estrogen, are disrupted, with well-established roles in bone metabolism (Table II; Fig. 4).
BCAA catabolism primarily occurs in skeletal muscle, where they are broken down into their respective ketoacids and further metabolized. This process is essential for maintaining muscle function and overall metabolic balance. Impaired BCAA catabolism can lead to elevated circulating levels of BCAAs and their ketoacids, which are associated with metabolic disturbances that may contribute to the development of osteoporosis. For example, increased BCAA levels can disrupt insulin signaling pathways, potentially resulting in insulin resistance, a recognized risk factor for osteoporosis (149).
Moreover, the regulation of key enzymes in BCAA metabolism, such as BCAT and BCKDH, is crucial for maintaining metabolic homeostasis. Inhibition of BCAT1 has been shown to impair osteoclast differentiation, suggesting its potential as a therapeutic target to halt the progression of osteoporosis (12). Emerging evidence has indicated that the expression of these metabolic enzymes is influenced by factors such as physical activity and transcriptional coactivators, including PGC-1α. In skeletal muscle, PGC-1α enhances the expression of BCAA-metabolizing enzymes, facilitating BCAA degradation and potentially alleviating the detrimental effects of their accumulation (150).
As the incidence of osteoporotic events continues to increase, the management of osteoporosis has increasingly focused on long-term strategies. Consequently, identifying safe, accessible and effective nutritional interventions to maintain skeletal health has become a key area of clinical research. BCAAs, due to their widespread presence in daily diets and ease of adherence, have attracted growing interest for their potential effects on bone metabolism, suggesting translational and clinical intervention value.
Cellular and animal studies have shown that exogenous BCAAs can modulate bone metabolism through the activation of signaling pathways, such as mTOR. This regulation occurs through two concurrent effects: Promoting osteogenic differentiation and inhibiting excessive osteoclast activation (151-154). Additionally, clinical observational studies have identified a positive association between higher BCAA levels and the preservation of bone mineral density (BMD) (151). These findings suggest that BCAA supplementation may offer therapeutic potential in delaying osteoporosis progression. Similarly, Carbone et al (155) and Urano et al (156) have indicated that higher BCAA levels are associated with better BMD. Together, these studies imply that elevated BCAA levels may be associated with improved bone health and a reduced risk of osteoporosis.
However, these findings do not establish a causal relationship. Residual confounding factors, such as total dietary protein intake, combination with other substances or physical activity, may influence both BCAA levels and bone health. Additionally, the relationship between BCAAs and bone health has been reported to vary across different disease stages or metabolic conditions (116). Therefore, well-designed clinical studies are critical, and further research involving additional human data is necessary.
Among the BCAAs, leucine is recognized as a critical amino acid for muscle function (157-159). BCAAs can effectively prevent and improve sarcopenia, as well as reduce fat infiltration in skeletal muscles, thus preserving muscle mass and function (160). A clinical intervention study conducted by Wang et al (161) demonstrated that leucine supplementation enhances muscle performance by improving cytoskeletal dynamics.
Skeletal muscle serves a pivotal role in bone growth and remodeling (162). Accumulating evidence has indicated that mechanical stimulation exerted by skeletal muscle promotes the proliferation and maturation of chondrocytes through factors such as osteocrin, which is secreted by periosteal osteoblasts, contributing to the elongation of long bones (163). Furthermore, mechanical signals regulate the dynamic balance between bone formation and resorption, enhancing bone strength through chemical messengers, including nitric oxide produced by osteocytes (164). Muscles also influence bones through the secretion of factors such as irisin, which promotes bone mass accumulation by inhibiting sclerostin expression, enhancing bone formation and reducing osteoclast numbers to suppress bone resorption (165). These mechanisms collectively impact the onset and progression of osteoporosis (166).
In elderly individuals and post-surgical rehabilitation populations, the combination of BCAA supplementation and exercise has proven effective in enhancing muscle strength, which is critical for restoring mobility and providing mechanical stimulation to the skeleton (167,168). Although the direct efficacy of BCAA supplementation in treating osteoporosis in humans requires further validation, it remains a promising approach and warrants targeted clinical research.
BCAT1 catalyzes the transfer of the amine group from BCAAs to α-ketoglutarate, generating glutamate and α-ketoacids, which are subsequently oxidized to provide cellular energy. Therefore, BCAT1 is crucial for preserving BCAA homeostasis and ensuring their continued availability for energy production. In a mouse model, Go et al (12) observed that a 400 μM BCAA solution effectively promoted osteoclast differentiation, whereas a higher concentration (800 μM) inhibited it, suggesting a negative feedback mechanism for BCAAs. Additionally, Huynh et al reported (169) that mTORC1, a key nutrient sensor known to be activated by BCAAs (98), inhibits osteoclast differentiation through calcineurin and NFATc1 signaling. BCAA levels are regulated not only by BCATs but also by their primary transporter, LAT1. Ozaki et al demonstrated that deleting the gene encoding LAT1 (Slc7a5) specifically in osteoclasts resulted in osteoclast activation and bone loss (170). Furthermore, Pereira et al (117) validated the functional role of the macrophage multinucleation network (MMnet), a trans-regulated gene co-expression network enriched for osteoclast-related genes, in regulating osteoclast multinucleation, resorption, bone mass and strength. These findings suggest that factors such as BCAT1, mTORC1, Slc7a5 and MMnet may serve as potential therapeutic targets to inhibit osteoclast formation, offering a strategy for osteoporosis treatment.
A recent study indicated that elevated BCAA levels are associated with insulin resistance and an increased risk of metabolic syndrome (171). In addition, a longitudinal study conducted between 2016 and 2022 with 3,090 Brazilian participants revealed a positive association between total BCAA intake (specifically >16 g) and the risk of obesity (172). These findings, along with the results from other experiments, are summarized in Table III. Additionally, in populations with BCAA metabolism disorders, such as patients with diabetes mellitus and liver cirrhosis, abnormal BCAA levels may impact bone metabolism (173). Therefore, the potential benefits of regulating BCAAs must be evaluated in conjunction with the metabolic health status of the individual.
Recently, You et al (174) developed alendronate-functionalized bone-targeting liposomes that encapsulate arginine and metformin. These liposomes enhance therapeutic efficacy against osteoporosis through dual mechanisms: Combined bone-targeted delivery and modulation of the hepatic-osseous axis. By integrating advanced nanomedicine technologies, the successful establishment of a multi-drug synergy strategy, featuring bone-targeted delivery and a collaborative mechanism, provides a novel approach to addressing potential adverse reactions in BCAA therapy. Additionally, personalized metabolic profiles derived from multi-omics analysis allow for the implementation of precise nutritional interventions. These interventions aim to identify the optimal BCAA intake level that supports bone health without inducing metabolic disorders, striking a key balance between therapeutic benefits and risks (175). However, further well-designed clinical studies are required to explore the relationship between BCAAs and bone metabolism/osteoporosis. Currently, notable scientific challenges, technical barriers and gaps in human data limit the clinical translation of BCAA therapy.
The accumulation of adipose tissue within the bone marrow space can induce an inflammatory state (176), which in turn promotes bone resorption and impairs the differentiation of MSCs and hematopoietic stem cells (177). Furthermore, plant-based foods, rich in dietary fiber and polyphenols, can enhance energy expenditure through thermogenesis and regulation of lipid metabolism, thereby helping reduce fat accumulation (178). Increasing the proportion of healthy plant-based foods in the diet, which may reduce bone marrow adipose tissue or modulate the function of bone marrow adipocytes, could serve as a potential target for improving bone quality and has important implications for the development of novel therapeutic strategies (179).
BCAA has attracted increasing attention due to its diverse biological activities and therapeutic potential. The present review focused on its effects in bone metabolism and osteoporosis. Notably, BCAA regulates the muscular and immune systems by activating the mTOR pathway, thereby influencing bone function. It also acts as a molecular signal to exert direct or indirect effects on bone metabolism and bone homeostasis. BCAA metabolic disorders contribute to a complex pathological network that involves not only the direct regulation of osteoblasts and osteoclasts, but also indirect effects mediated by systemic metabolic abnormalities. Collectively, these factors disrupt bone homeostasis and may ultimately lead to osteoporosis.
Evidence from animal and human studies have suggested that BCAA intake may be associated with increased BMD, supporting its potential as a nutritional target for maintaining bone health and preventing osteoporosis; however, most current evidence is derived from in vitro and animal experiments, which are limited by interspecies metabolic differences and dose conversion issues, restricting clinical translation (180,181). Moreover, high-quality population-based studies and prospective intervention trials remain scarce. Existing studies often suffer from poor reproducibility due to small sample sizes and inconsistent intervention methods (182,183). The specific mechanisms of BCAA action within the bone microenvironment have not been fully elucidated, and the effects of individual metabolic differences and disease states on BCAA metabolism require further investigation. Future work should explore the molecular mechanisms of BCAA in greater depth. Multicenter, large-sample, long-term randomized controlled trials are needed to determine the long-term effects of different BCAA doses and ratios on BMD and bone turnover markers (155). Integration of advanced technologies, such as metabolomics, may promote the development of precise nutritional intervention strategies.
As research on the modulation of bone metabolism by BCAAs and their role in osteoporosis treatment continues to grow, these studies will further enhance the understanding and improve strategies targeting BCAA metabolic processes in disease therapy. The current review not only provides a reference for further development and investigation of the impact of BCAA metabolism on osteoporosis progression, but also serves as a guide for regulating BCAA metabolism to achieve precision treatment of osteoporosis. Furthermore, these targeted therapeutic strategies may offer innovative treatment options for bone metabolism-related diseases.
Not applicable.
QX, HZ and RY drafted and revised the manuscript. YZ and FL contributed to the literature search, evidence extraction, thematic organization, critical interpretation of the reviewed studies, and the preparation and refinement of the figures. BC contributed to the literature search, screening, evidence evaluation, and assisted in organizing and interpreting the included studies. XC conceived the review, supervised the overall academic direction and critically revised the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
BCAA |
branched-chain amino acid |
|
RANKL |
receptor activator of NF-κB ligand |
|
RANK |
receptor activator of NF-κB |
|
TGF-β |
transforming growth factor-β |
|
IGF-1 |
insulin-like growth factor-1 |
|
ATP |
adenosine triphosphate |
|
PANX1 |
Pannexin 1 |
|
P2X7R |
P2X7 receptor |
|
OPG |
osteoprotegerin |
|
PTH |
parathyroid hormone |
|
Cx43 |
connexin 43 |
|
BCAT |
BCAA transaminase |
|
BCKDH |
branched-chain α-ketoacid dehydrogenase |
|
BCKAs |
branched-chain α-ketoacids |
|
mTOR |
mechanistic target of rapamycin |
|
mTORC |
mTOR complex |
|
BAIBA |
β-aminoisobutyric acid |
|
3-HIB |
3-hydroxyisobutyric acid |
|
PGC-1α |
peroxisome proliferator-activated receptor γ coactivator-1α |
|
TM |
memory T |
|
TEM |
effector TM |
|
BMSCs |
bone marrow mesenchymal stem cells |
|
NAP1L2 |
nucleosome assembly protein 1 like 2 |
|
BMD |
bone mineral density |
|
MMnet |
macrophage multinucleation network |
|
DAMP |
damage-associated molecular pattern |
|
PRRs |
pattern recognition receptors |
|
P2Y2 |
purinergic receptor P2Y 2 |
|
P2Y1 |
purinergic receptor P2Y 1 |
|
PTHrP |
PTH-related protein |
|
GH |
growth hormone |
|
cAMP |
cyclic adenosine monophosphate |
|
PKA |
protein kinase A |
|
KIC |
α-ketoisocaproate |
|
KIV |
α-ketoisovalerate |
|
KMA |
α-ketomethylvalerate |
|
PTH1R |
PTH1 receptor |
|
TCA |
tricarboxylic acid |
|
α-KG |
α-ketoglutarate |
|
BMDCs |
bone marrow dendritic cells |
|
SASP |
senescence-associated secretory phenotype |
|
Th17 |
T helper 17 |
Not applicable.
The present review was supported by the National Natural Science Foundation of China Incubation Project (grant no. Institute-level: 2023YNFY12009); the National College Students' Innovative Entrepreneurial Training Plan Program (grant no. 202410403067); the Innovation and Entrepreneurship Training Program for College Students in Jiangxi Province (grant no. S202410403035); the Doctoral Start-up Fund (grant no. B3477); and the Nanchang University Second Affiliated Hospital In-Hospital Project (grant no. 2023efy002).
|
Li H, Xiao Z, Quarles LD and Li W: Osteoporosis: Mechanism, molecular target and current status on drug development. Curr Med Chem. 28:1489–1507. 2021. View Article : Google Scholar | |
|
Zhang X, Wang Z, Zhang D, Ye D, Zhou Y, Qin J and Zhang Y: The prevalence and treatment rate trends of osteoporosis in postmenopausal women. PLoS One. 18:e02902892023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Yu W, Yin X, Cui L, Tang S, Jiang N, Cui L, Zhao N, Lin Q, Chen L, et al: Prevalence of osteoporosis and fracture in China: The China osteoporosis prevalence study. JAMA Netw Open. 4:e21211062021. View Article : Google Scholar : PubMed/NCBI | |
|
Center JR, Nguyen TV, Schneider D, Sambrook PN and Eisman JA: Mortality after all major types of osteoporotic fracture in men and women: An observational study. Lancet. 353:878–882. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Harada S and Rodan GA: Control of osteoblast function and regulation of bone mass. Nature. 423:349–355. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Raisz LG: Pathogenesis of osteoporosis: Concepts, conflicts, and prospects. J Clin Invest. 115:3318–3325. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Gao M, Gao W, Papadimitriou JM, Zhang C, Gao J and Zheng M: Exosomes-the enigmatic regulators of bone homeostasis. Bone Res. 6:362018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Li Q, Peng X, Ji P, Zhang Y, Jin J, Yuan Z, Jiang J, Tian G, Cai M, et al: Targeting chaperone-mediated autophagy to regulate osteoclast activity as a therapeutic strategy for osteoporosis. Mater Today Bio. 35:1023112025. View Article : Google Scholar : PubMed/NCBI | |
|
Harper AE, Miller RH and Block KP: Branched-chain amino acid metabolism. Annu Rev Nutr. 4:409–454. 1984. View Article : Google Scholar : PubMed/NCBI | |
|
Mansoori S, Ho MY, Ng KK and Cheng KK: Branched-chain amino acid metabolism: Pathophysiological mechanism and therapeutic intervention in metabolic diseases. Obes Rev. 26:e138562025. View Article : Google Scholar | |
|
Trautman ME, Richardson NE and Lamming DW: Protein restriction and branched-chain amino acid restriction promote geroprotective shifts in metabolism. Aging Cell. 21:e136262022. View Article : Google Scholar : PubMed/NCBI | |
|
Go M, Shin E, Jang SY, Nam M, Hwang GS and Lee SY: BCAT1 promotes osteoclast maturation by regulating branched-chain amino acid metabolism. Exp Mol Med. 54:825–833. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Weiner S and Traub W: Bone structure: From angstroms to microns. FASEB J. 6:879–885. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Hua R, Han Y, Ni Q, Fajardo RJ, Iozzo RV, Ahmed R, Nyman JS, Wang X and Jiang JX: Pivotal roles of biglycan and decorin in regulating bone mass, water retention, and bone toughness. Bone Res. 13:22025. View Article : Google Scholar : PubMed/NCBI | |
|
Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, et al: Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 425:841–846. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, et al: Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 425:836–841. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Taichman RS and Emerson SG: Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med. 179:1677–1682. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Bolamperti S, Villa I and Rubinacci A: Bone remodeling: an operational process ensuring survival and bone mechanical competence. Bone Res. 10:482022. View Article : Google Scholar : PubMed/NCBI | |
|
Hattner R, Epker BN and Frost HM: Suggested sequential mode of control of changes in cell behaviour in adult bone remodelling. Nature. 206:489–490. 1965. View Article : Google Scholar : PubMed/NCBI | |
|
Barak MM: Bone modeling or bone remodeling: That is the question. Am J Phys Anthropol. 172:153–155. 2020. View Article : Google Scholar | |
|
Yan C, Zhang P, Qin Q, Jiang K, Luo Y, Xiang C, He J, Chen L, Jiang D, Cui W and Li Y: 3D-printed bone regeneration scaffolds modulate bone metabolic homeostasis through vascularization for osteoporotic bone defects. Biomaterials. 311:1226992024. View Article : Google Scholar : PubMed/NCBI | |
|
Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishihara T, Koga T, Martin TJ and Suda T: Origin of osteoclasts: mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells. Proc Natl Acad Sci USA. 87:7260–7264. 1990. View Article : Google Scholar : PubMed/NCBI | |
|
Boyle WJ, Simonet WS and Lacey DL: Osteoclast differentiation and activation. Nature. 423:337–342. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Teitelbaum SL: Bone resorption by osteoclasts. Science. 289:1504–1508. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Nakahama KI, Hidaka S, Goto K, Tada M, Doi T, Nakamura H, Akiyama M and Shinohara M: Visualization and quantification of RANK-RANKL binding for application to disease investigations and drug discovery. Bone. 195:1174732025. View Article : Google Scholar : PubMed/NCBI | |
|
Holliday LS, Patel SS and Rody WJ Jr: RANKL and RANK in extracellular vesicles: Surprising new players in bone remodeling. Extracell Vesicles Circ Nucl Acids. 2:18–28. 2021.PubMed/NCBI | |
|
Soriano P, Montgomery C, Geske R and Bradley A: Targeted disruption of the c-src proto-oncogene leads to osteopetrosis in mice. Cell. 64:693–702. 1991. View Article : Google Scholar : PubMed/NCBI | |
|
Boyce BF, Yoneda T, Lowe C, Soriano P and Mundy GR: Requirement of pp60c-src expression for osteoclasts to form ruffled borders and resorb bone in mice. J Clin Invest. 90:1622–1627. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Gelb BD, Shi GP, Chapman HA and Desnick RJ: Pycnodysostosis, a lysosomal disease caused by cathepsin K deficiency. Science. 273:1236–1238. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Saftig P, Hunziker E, Everts V, Jones S, Boyde A, Wehmeyer O, Suter A and von Figura K: Functions of cathepsin K in bone resorption. Lessons from cathepsin K deficient mice. Adv Exp Med Biol. 477:293–303. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Mizoguchi T and Ono N: The diverse origin of bone-forming osteoblasts. J Bone Miner Res. 36:1432–1447. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, Zhao L, Nagy TR, Peng X, Hu J, et al: TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med. 15:757–765. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Bodine PV and Komm BS: Wnt signaling and osteoblastogenesis. Rev Endocr Metab Disord. 7:33–39. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Eriksen EF: Cellular mechanisms of bone remodeling. Rev Endocr Metab Disord. 11:219–227. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Hong AR, Kim K, Lee JY, Yang JY, Kim JH, Shin CS and Kim SW: Transformation of mature osteoblasts into bone lining cells and RNA sequencing-based transcriptome profiling of mouse bone during mechanical unloading. Endocrinol Metab (Seoul). 35:456–469. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang JS and Wein MN: Pathways controlling formation and maintenance of the osteocyte dendrite network. Curr Osteoporos Rep. 20:493–504. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Komori T: Cell death in chondrocytes, osteoblasts, and osteocytes. Int J Mol Sci. 17:20452016. View Article : Google Scholar : PubMed/NCBI | |
|
Takeuchi O and Akira S: Pattern recognition receptors and inflammation. Cell. 140:805–820. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, Tracey KJ and Chiorazzi N: High mobility group box protein 1: An endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol. 173:307–313. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Giannoni P, Marini C, Cutrona G, Matis S, Capra MC, Puglisi F, Luzzi P, Pigozzi S, Gaggero G, Neri A, et al: Chronic lymphocytic leukemia cells impair osteoblastogenesis and promote osteoclastogenesis: Role of TNFα, IL-6 and IL-11 cytokines. Haematologica. 106:2598–2612. 2021. View Article : Google Scholar | |
|
Kringelbach TM, Aslan D, Novak I, Schwarz P and Jørgensen NR: UTP-induced ATP release is a fine-tuned signalling pathway in osteocytes. Purinergic Signal. 10:337–347. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Verborgt O, Gibson GJ and Schaffler MB: Loss of osteocyte integrity in association with microdamage and bone remodeling after fatigue in vivo. J Bone Miner Res. 15:60–67. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Cheung WY, Fritton JC, Morgan SA, Seref-Ferlengez Z, Basta-Pljakic J, Thi MM, Suadicani SO, Spray DC, Majeska RJ and Schaffler MB: Pannexin-1 and P2X7-receptor are required for apoptotic osteocytes in fatigued bone to trigger RANKL production in neighboring bystander osteocytes. J Bone Miner Res. 31:890–899. 2016. View Article : Google Scholar | |
|
Luckprom P, Wongkhantee S, Yongchaitrakul T and Pavasant P: Adenosine triphosphate stimulates RANKL expression through P2Y1 receptor-cyclo-oxygenase-dependent pathway in human periodontal ligament cells. J Periodontal Res. 45:404–411. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Buckley KA, Hipskind RA, Gartland A, Bowler WB and Gallagher JA: Adenosine triphosphate stimulates human osteoclast activity via upregulation of osteoblast-expressed receptor activator of nuclear factor-kappa B ligand. Bone. 31:582–590. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Gallagher JA: ATP P2 receptors and regulation of bone effector cells. J Musculoskelet Neuronal Interact. 4:125–127. 2004.PubMed/NCBI | |
|
Cusato K, Bosco A, Rozental R, Guimarães CA, Reese BE, Linden R and Spray DC: Gap junctions mediate bystander cell death in developing retina. J Neurosci. 23:6413–6422. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Cawley KM, Bustamante-Gomez NC, Guha AG, MacLeod RS, Xiong J, Gubrij I, Liu Y, Mulkey R, Palmieri M, Thostenson JD, et al: Local production of osteoprotegerin by osteoblasts suppresses bone resorption. Cell Rep. 32:1080522020. View Article : Google Scholar : PubMed/NCBI | |
|
Kennedy OD, Laudier DM, Majeska RJ, Sun HB and Schaffler MB: Osteocyte apoptosis is required for production of osteoclastogenic signals following bone fatigue in vivo. Bone. 64:132–137. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Verborgt O, Tatton NA, Majeska RJ and Schaffler MB: Spatial distribution of Bax and Bcl-2 in osteocytes after bone fatigue: complementary roles in bone remodeling regulation? J Bone Miner Res. 17:907–914. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Lieben L, Carmeliet G and Masuyama R: Calcemic actions of vitamin D: Effects on the intestine, kidney and bone. Best Pract Res Clin Endocrinol Metab. 25:561–572. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Zarei A, Morovat A, Javaid K and Brown CP: Vitamin D receptor expression in human bone tissue and dose-dependent activation in resorbing osteoclasts. Bone Res. 4:160302016. View Article : Google Scholar : PubMed/NCBI | |
|
Lips P and van Schoor NM: The effect of vitamin D on bone and osteoporosis. Best Pract Res Clin Endocrinol Metab. 25:585–591. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Bikle DD: Vitamin D and bone. Curr Osteoporos Rep. 10:151–159. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Arnaud CD Jr, Tenenhouse AM and Rasmussen H: Parathyroid hormone. Annu Rev Physiol. 29:349–372. 1967. View Article : Google Scholar : PubMed/NCBI | |
|
Martin TJ, Sims NA and Seeman E: Physiological and pharmacological roles of PTH and PTHrP in bone using their shared receptor, PTH1R. Endocr Rev. 42:383–406. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Quarles LD: Skeletal secretion of FGF-23 regulates phosphate and vitamin D metabolism. Nat Rev Endocrinol. 8:276–286. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Jüppner H: Phosphate and FGF-23. Kidney Int. 79121:S24–S27. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Naot D, Musson DS and Cornish J: The activity of peptides of the calcitonin family in bone. Physiol Rev. 99:781–805. 2019. View Article : Google Scholar | |
|
Xu J, Wang J, Chen X, Li Y, Mi J and Qin L: The effects of calcitonin gene-related peptide on bone homeostasis and regeneration. Curr Osteoporos Rep. 18:621–632. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lecanda F, Warlow PM, Sheikh S, Furlan F, Steinberg TH and Civitelli R: Connexin43 deficiency causes delayed ossification, craniofacial abnormalities, and osteoblast dysfunction. J Cell Biol. 151:931–944. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Chung DJ, Castro CH, Watkins M, Stains JP, Chung MY, Szejnfeld VL, Willecke K, Theis M and Civitelli R: Low peak bone mass and attenuated anabolic response to parathyroid hormone in mice with an osteoblast-specific deletion of connexin43. J Cell Sci. 119(Pt 20): 4187–4198. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Paul EM, Sathyendra V, Davison A, Sharkey N, Bronson S, Srinivasan S, Gross TS and Donahue HJ: Enhanced osteoclastic resorption and responsiveness to mechanical load in gap junction deficient bone. PLoS One. 6:e235162011. View Article : Google Scholar : PubMed/NCBI | |
|
Tiede-Lewis LM, Xie Y, Hulbert MA, Campos R, Dallas MR, Dusevich V, Bonewald LF and Dallas SL: Degeneration of the osteocyte network in the C57BL/6 mouse model of aging. Aging (Albany NY). 9:2190–2208. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dole NS, Mazur CM, Acevedo C, Lopez JP, Monteiro DA, Fowler TW, Gludovatz B, Walsh F, Regan JN, Messina S, et al: Osteocyte-Intrinsic TGF-β signaling regulates bone quality through perilacunar/canalicular remodeling. Cell Rep. 21:2585–2596. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kreis NN, Friemel A, Ritter A, Hentrich AE, Siebelitz E, Louwen F and Yuan J: In-depth analysis of obesity-associated changes in adipose tissue-derived mesenchymal stromal/stem cells and primary cilia function. Commun Biol. 8:14622025. View Article : Google Scholar : PubMed/NCBI | |
|
Olney RC: Regulation of bone mass by growth hormone. Med Pediatr Oncol. 41:228–234. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Iglesias L, Yeh JK, Castro-Magana M and Aloia JF: Effects of growth hormone on bone modeling and remodeling in hypophysectomized young female rats: A bone histomorphometric study. J Bone Miner Metab. 29:159–167. 2011. View Article : Google Scholar | |
|
Hayashi M, Nakashima T, Yoshimura N, Okamoto K, Tanaka S and Takayanagi H: Autoregulation of osteocyte Sema3A orchestrates estrogen action and counteracts bone aging. Cell Metab. 29:627–637.e5. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma D, Larriera AI, Palacio-Mancheno PE, Gatti V, Fritton JC, Bromage TG, Cardoso L, Doty SB and Fritton SP: The effects of estrogen deficiency on cortical bone microporosity and mineralization. Bone. 110:1–10. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jackson E, Lara-Castillo N, Akhter MP, Dallas M, Scott JM, Ganesh T and Johnson ML: Osteocyte Wnt/β-catenin pathway activation upon mechanical loading is altered in ovariectomized mice. Bone Rep. 15:1011292021. View Article : Google Scholar | |
|
Ma L, Hua R, Tian Y, Cheng H, Fajardo RJ, Pearson JJ, Guda T, Shropshire DB, Gu S and Jiang JX: Connexin 43 hemichannels protect bone loss during estrogen deficiency. Bone Res. 7:112019. View Article : Google Scholar : PubMed/NCBI | |
|
He Z, Li H, Han X, Zhou F, Du J, Yang Y, Xu Q, Zhang S, Zhang S, Zhao N, et al: Irisin inhibits osteocyte apoptosis by activating the Erk signaling pathway in vitro and attenuates ALCT-induced osteoarthritis in mice. Bone. 141:1155732020. View Article : Google Scholar : PubMed/NCBI | |
|
Al-Suhaimi EA and Al-Jafary MA: Endocrine roles of vitamin K-dependent-osteocalcin in the relation between bone metabolism and metabolic disorders. Rev Endocr Metab Disord. 21:117–125. 2020. View Article : Google Scholar | |
|
Yang D, Gong G, Song J, Chen J, Wang S, Li J and Wang G: Ferroptosis-mediated osteoclast-osteoblast crosstalk: Signaling pathways governing bone remodeling in osteoporosis. J Orthop Surg Res. 20:8882025. View Article : Google Scholar : PubMed/NCBI | |
|
Baek KH, Chung YS, Koh JM, Kim IJ, Kim KM, Min YK, Park KD, Dinavahi R, Maddox J, Yang W, et al: Romosozumab in postmenopausal Korean women with osteoporosis: A Randomized, double-blind, placebo-controlled efficacy and safety study. Endocrinol Metab (Seoul). 36:60–69. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kasiske BL, Kumar R, Kimmel PL, Pesavento TE, Kalil RS, Kraus ES, Rabb H, Posselt AM, Anderson-Haag TL, Steffes MW, et al: Abnormalities in biomarkers of mineral and bone metabolism in kidney donors. Kidney Int. 90:861–868. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Schwetz V, Pieber T and Obermayer-Pietsch B: The endocrine role of the skeleton: Background and clinical evidence. Eur J Endocrinol. 166:959–967. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kärkkäinen M, Tuppurainen M, Salovaara K, Sandini L, Rikkonen T, Sirola J, Honkanen R, Jurvelin J, Alhava E and Kröger H: Effect of calcium and vitamin D supplementation on bone mineral density in women aged 65-71 years: A 3-year randomized population-based trial (OSTPRE-FPS). Osteoporos Int. 21:2047–2055. 2010. View Article : Google Scholar | |
|
Remer T, Krupp D and Shi L: Dietary protein's and dietary acid load's influence on bone health. Crit Rev Food Sci Nutr. 54:1140–1150. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Langeveld M and Hollak CEM: Bone health in patients with inborn errors of metabolism. Rev Endocr Metab Disord. 19:81–92. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yang K, Li J and Tao L: Purine metabolism in the development of osteoporosis. Biomed Pharmacother. 155:1137842022. View Article : Google Scholar : PubMed/NCBI | |
|
Iantomasi T, Romagnoli C, Palmini G, Donati S, Falsetti I, Miglietta F, Aurilia C, Marini F, Giusti F and Brandi ML: Oxidative stress and inflammation in osteoporosis: Molecular mechanisms involved and the relationship with microRNAs. Int J Mol Sci. 24:37722023. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson JT, Hussain MA, Cherian KE, Kapoor N and Paul TV: Chronic alcohol consumption and its impact on bone and metabolic health - A narrative review. Indian J Endocrinol Metab. 26:206–212. 2022.PubMed/NCBI | |
|
Ehnert S, Aspera-Werz RH, Ihle C, Trost M, Zirn B, Flesch I, Schröter S, Relja B and Nussler AK: Smoking dependent alterations in bone formation and inflammation represent major risk factors for complications following total joint arthroplasty. J Clin Med. 8:4062019. View Article : Google Scholar : PubMed/NCBI | |
|
Huang YS, Gao JW, Ao RF, Liu XY, Wu DZ, Huang JL, Tu C, Zhuang JS, Zhu SY and Zhong ZM: Accumulation of advanced oxidation protein products aggravates bone-fat imbalance during skeletal aging. J Orthop Translat. 51:24–36. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Schröder K: NADPH oxidases in bone homeostasis and osteoporosis. Free Radic Biol Med. 132:67–72. 2019. View Article : Google Scholar | |
|
Jiang N, Liu J, Guan C, Ma C, An J and Tang X: Thioredoxin-interacting protein: A new therapeutic target in bone metabolism disorders? Front Immunol. 13:9551282022. View Article : Google Scholar : PubMed/NCBI | |
|
Le Couteur DG, Solon-Biet SM, Cogger VC, Ribeiro R, de Cabo R, Raubenheimer D, Cooney GJ and Simpson SJ: Branched chain amino acids, aging and age-related health. Ageing Res Rev. 64:1011982020. View Article : Google Scholar : PubMed/NCBI | |
|
Brosnan JT and Brosnan ME: Branched-chain amino acids: Enzyme and substrate regulation. J Nutr. 136(1 Suppl): 207S–211S. 2006. View Article : Google Scholar | |
|
Han JM, Jeong SJ, Park MC, Kim G, Kwon NH, Kim HK, Ha SH, Ryu SH and Kim S: Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell. 149:410–424. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang S, Zeng X, Ren M, Mao X and Qiao S: Novel metabolic and physiological functions of branched chain amino acids: A review. J Anim Sci Biotechnol. 8:102017. View Article : Google Scholar : PubMed/NCBI | |
|
Neinast MD, Jang C, Hui S, Murashige DS, Chu Q, Morscher RJ, Li X, Zhan L, White E, Anthony TG, et al: Quantitative analysis of the whole-body metabolic fate of branched-chain amino acids. Cell Metab. 29:417–429.e4. 2019. View Article : Google Scholar | |
|
Wang W, Liu Z, Liu L, Han T, Yang X and Sun C: Genetic predisposition to impaired metabolism of the branched chain amino acids, dietary intakes, and risk of type 2 diabetes. Genes Nutr. 16:202021. View Article : Google Scholar : PubMed/NCBI | |
|
Bonvini A, Coqueiro AY, Tirapegui J, Calder PC and Rogero MM: Immunomodulatory role of branched-chain amino acids. Nutr Rev. 76:840–856. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Deng AF, Wang FX, Wang SC, Zhang YZ, Bai L and Su JC: Bone-organ axes: Bidirectional crosstalk. Mil Med Res. 11:372024.PubMed/NCBI | |
|
Hasegawa T, Kikuta J, Sudo T, Matsuura Y, Matsui T, Simmons S, Ebina K, Hirao M, Okuzaki D, Yoshida Y, et al: Identification of a novel arthritis-associated osteoclast precursor macrophage regulated by FoxM1. Nat Immunol. 20:1631–1643. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Anthony JC, Yoshizawa F, Anthony TG, Vary TC, Jefferson LS and Kimball SR: Leucine stimulates translation initiation in skeletal muscle of postabsorptive rats via a rapamycin-sensitive pathway. J Nutr. 130:2413–2419. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Yang M, Zhang X, Ding Y, Yang L, Ren W, Gao Y, Yao K, Zhou Y and Shao W: The effect of valine on the synthesis of α-Casein in MAC-T cells and the expression and phosphorylation of genes related to the mTOR signaling pathway. Int J Mol Sci. 26:31792025. View Article : Google Scholar | |
|
Glantschnig H, Fisher JE, Wesolowski G, Rodan GA and Reszka AA: M-CSF, TNFalpha and RANK ligand promote osteoclast survival by signaling through mTOR/S6 kinase. Cell Death Differ. 10:1165–1177. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Kneissel M, Luong-Nguyen NH, Baptist M, Cortesi R, Zumstein-Mecker S, Kossida S, O'Reilly T, Lane H and Susa M: Everolimus suppresses cancellous bone loss, bone resorption, and cathepsin K expression by osteoclasts. Bone. 35:1144–1156. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Ory B, Moriceau G, Redini F and Heymann D: mTOR inhibitors (rapamycin and its derivatives) and nitrogen containing bisphosphonates: Bi-functional compounds for the treatment of bone tumours. Curr Med Chem. 14:1381–1387. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Benslimane-Ahmim Z, Heymann D, Dizier B, Lokajczyk A, Brion R, Laurendeau I, Bièche I, Smadja DM, Galy-Fauroux I, Colliec-Jouault S, et al: Osteoprotegerin, a new actor in vasculogenesis, stimulates endothelial colony-forming cells properties. J Thromb Haemost. 9:834–843. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Gnant M, Baselga J, Rugo HS, Noguchi S, Burris HA, Piccart M, Hortobagyi GN, Eakle J, Mukai H, Iwata H, et al: Effect of everolimus on bone marker levels and progressive disease in bone in BOLERO-2. J Natl Cancer Inst. 105:654–663. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Roberts LD, Boström P, O'Sullivan JF, Schinzel RT, Lewis GD, Dejam A, Lee YK, Palma MJ, Calhoun S, Georgiadi A, et al: β-Aminoisobutyric acid induces browning of white fat and hepatic β-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 19:96–108. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Jang C, Oh SF, Wada S, Rowe GC, Liu L, Chan MC, Rhee J, Hoshino A, Kim B, Ibrahim A, et al: A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance. Nat Med. 22:421–426. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Batch BC, Shah SH, Newgard CB, Turer CB, Haynes C, Bain JR, Muehlbauer M, Patel MJ, Stevens RD, Appel LJ, et al: Branched chain amino acids are novel biomarkers for discrimination of metabolic wellness. Metabolism. 62:961–969. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Connelly MA, Wolak-Dinsmore J and Dullaart RPF: Branched chain amino acids are associated with insulin resistance independent of leptin and adiponectin in subjects with varying degrees of glucose tolerance. Metab Syndr Relat Disord. 15:183–186. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Lotta LA, Scott RA, Sharp SJ, Burgess S, Luan J, Tillin T, Schmidt AF, Imamura F, Stewart ID, Perry JR, et al: Genetic predisposition to an impaired metabolism of the branched-chain amino acids and risk of type 2 diabetes: A mendelian randomisation analysis. PLoS Med. 13:e10021792016. View Article : Google Scholar : PubMed/NCBI | |
|
Felig P, Marliss E and Cahill GF Jr: Plasma amino acid levels and insulin secretion in obesity. N Engl J Med. 281:811–816. 1969. View Article : Google Scholar : PubMed/NCBI | |
|
Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA, et al: A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 9:311–326. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Holmes MV, Davey Smith G and Ala-Korpela M: Genetic support for a causal role of insulin resistance on circulating branched-chain amino acids and inflammation. Diabetes Care. 40:1779–1786. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mahendran Y, Jonsson A, Have CT, Allin KH, Witte DR, Jørgensen ME, Grarup N, Pedersen O, Kilpeläinen TO and Hansen T: Genetic evidence of a causal effect of insulin resistance on branched-chain amino acid levels. Diabetologia. 60:873–878. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Lynch CJ and Adams SH: Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol. 10:723–736. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Z, Su Z, Yang H, Zheng J, Wu X, Huang M, Duan L, Chen S, Wei B, Fan X and Lin S: Branched-chain amino acids in bone health: From molecular mechanisms to therapeutic potential. Biomed Pharmacother. 192:1186452025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao H, Zhang F, Sun D, Wang X, Zhang X, Zhang J, Yan F, Huang C, Xie H, Lin C, et al: Branched-Chain amino acids exacerbate obesity-related hepatic glucose and lipid metabolic disorders via attenuating Akt2 signaling. Diabetes. 69:1164–1177. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pereira M, Ko JH, Logan J, Protheroe H, Kim KB, Tan ALM, Croucher PI, Park KS, Rotival M, Petretto E, et al: A trans-eQTL network regulates osteoclast multinucleation and bone mass. Elife. 9:e555492020. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JH and Kim N: Regulation of NFATc1 in osteoclast differentiation. J Bone Metab. 21:233–241. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yagi M, Miyamoto T, Sawatani Y, Iwamoto K, Hosogane N, Fujita N, Morita K, Ninomiya K, Suzuki T, Miyamoto K, et al: DC-STAMP is essential for cell-cell fusion in osteoclasts and foreign body giant cells. J Exp Med. 2005. 202:345–351. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Ishii M, Iwai K, Koike M, Ohshima S, Kudo-Tanaka E, Ishii T, Mima T, Katada Y, Miyatake K, Uchiyama Y and Saeki Y: RANKL-induced expression of tetraspanin CD9 in lipid raft membrane microdomain is essential for cell fusion during osteoclastogenesis. J Bone Miner Res. 21:965–976. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Singha UK, Jiang Y, Yu S, Luo M, Lu Y, Zhang J and Xiao G: Rapamycin inhibits osteoblast proliferation and differentiation in MC3T3-E1 cells and primary mouse bone marrow stromal cells. J Cell Biochem. 103:434–446. 2008. View Article : Google Scholar | |
|
Han HS, Ahn E, Park ES, Huh T, Choi S, Kwon Y, Choi BH, Lee J, Choi YH, Jeong YL, et al: Impaired BCAA catabolism in adipose tissues promotes age-associated metabolic derangement. Nat Aging. 3:982–1000. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen A, Jin J, Cheng S, Liu Z, Yang C, Chen Q, Liang W, Li K, Kang D, Ouyang Z, et al: mTORC1 induces plasma membrane depolarization and promotes preosteoblast senescence by regulating the sodium channel Scn1a. Bone Res. 10:252022. View Article : Google Scholar : PubMed/NCBI | |
|
Gayatri MB, Gajula NN, Chava S and Reddy ABM: High glutamine suppresses osteogenesis through mTORC1-mediated inhibition of the mTORC2/AKT-473/RUNX2 axis. Cell Death Discov. 8:2772022. View Article : Google Scholar : PubMed/NCBI | |
|
Arany Z and Neinast M: Branched chain amino acids in metabolic disease. Curr Diab Rep. 18:762018. View Article : Google Scholar : PubMed/NCBI | |
|
Yoon MS: The emerging role of branched-chain amino acids in insulin resistance and metabolism. Nutrients. 8:4052016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou M, Shao J, Wu CY, Shu L, Dong W, Liu Y, Chen M, Wynn RM, Wang J, Wang J, et al: Targeting BCAA Catabolism to Treat Obesity-Associated Insulin Resistance. Diabetes. 68:1730–1746. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou C, Zhang J, Liu Q, Guo Y, Li M, Tao J, Peng S, Li R, Deng X, Zhang G and Liu H: Role of amino acid metabolism in osteoporosis: Effects on the bone microenvironment and treatment strategies (Review). Mol Med Rep. 32:2122025. View Article : Google Scholar : PubMed/NCBI | |
|
Lerin C, Goldfine AB, Boes T, Liu M, Kasif S, Dreyfuss JM, De Sousa-Coelho AL, Daher G, Manoli I, Sysol JR, et al: Defects in muscle branched-chain amino acid oxidation contribute to impaired lipid metabolism. Mol Metab. 5:926–936. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
White PJ, Lapworth AL, An J, Wang L, McGarrah RW, Stevens RD, Ilkayeva O, George T, Muehlbauer MJ, Bain JR, et al: Branched-chain amino acid restriction in Zucker-fatty rats improves muscle insulin sensitivity by enhancing efficiency of fatty acid oxidation and acyl-glycine export. Mol Metab. 5:538–551. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Jian H, Xu Q, Wang X, Liu Y, Miao S, Li Y, Mou T, Dong X and Zou X: Amino acid and fatty acid metabolism disorders trigger oxidative stress and inflammatory response in excessive dietary valine-induced NAFLD of laying hens. Front Nutr. 9:8497672022. View Article : Google Scholar : PubMed/NCBI | |
|
Ye Z, Wang S, Zhang C and Zhao Y: Coordinated modulation of energy metabolism and inflammation by branched-chain amino acids and fatty acids. Front Endocrinol (Lausanne). 11:6172020. View Article : Google Scholar : PubMed/NCBI | |
|
Surugihalli C, Muralidaran V, Ryan CE, Patel K, Zhao D and Sunny NE: Branched-chain amino acids alter cellular redox to induce lipid oxidation and reduce de novo lipogenesis in the liver. Am J Physiol Endocrinol Metab. 324:E299–E313. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin S, Miao Y, Zheng X, Dong Y, Yang Q, Yang Q, Du S, Xu J, Zhou S and Yuan T: ANGPTL4 negatively regulates the progression of osteosarcoma by remodeling branched-chain amino acid metabolism. Cell Death Discov. 8:2252022. View Article : Google Scholar : PubMed/NCBI | |
|
Rachner TD, Khosla S and Hofbauer LC: Osteoporosis: Now and the future. Lancet. 377:1276–1287. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Riggs BL, Wahner HW, Seeman E, Offord KP, Dunn WL, Mazess RB, Johnson KA and Melton LJ III: Changes in bone mineral density of the proximal femur and spine with aging. Differences between the postmenopausal and senile osteoporosis syndromes. J Clin Invest. 70:716–723. 1982. View Article : Google Scholar : PubMed/NCBI | |
|
Wang LT, Chen LR and Chen KH: Hormone-related and drug-induced osteoporosis: A cellular and molecular overview. Int J Mol Sci. 24:58142023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu D, Cline-Smith A, Shashkova E, Perla A, Katyal A and Aurora R: T-cell mediated inflammation in postmenopausal osteoporosis. Front Immunol. 12:6875512021. View Article : Google Scholar : PubMed/NCBI | |
|
Bruyère O, Malaise O, Neuprez A, Collette J and Reginster JY: Prevalence of vitamin D inadequacy in European postmenopausal women. Curr Med Res Opin. 23:1939–1944. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Lee YH, Lim YW, Ling PS, Tan YY, Cheong M and Lam KS: Inadequate dietary calcium intake in elderly patients with hip fractures. Singapore Med J. 48:1117–1121. 2007.PubMed/NCBI | |
|
Nieves JW: Osteoporosis: The role of micronutrients. Am J Clin Nutr. 81:1232S–1239S. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Hu M, Xing L, Zhang L, Liu F, Wang S, Xie Y, Wang J, Jiang H, Guo J, Li X, et al: NAP1L2 drives mesenchymal stem cell senescence and suppresses osteogenic differentiation. Aging Cell. 21:e135512022. View Article : Google Scholar : PubMed/NCBI | |
|
Xue Y, Hu S, Chen C, He J, Sun J, Jin Y, Zhang Y, Zhu G, Shi Q and Rui Y: Myokine Irisin promotes osteogenesis by activating BMP/SMAD signaling via αV integrin and regulates bone mass in mice. Int J Biol Sci. 18:572–584. 2022. View Article : Google Scholar : | |
|
Mi B, Yan C, Xue H, Chen L, Panayi AC, Hu L, Hu Y, Cao F, Sun Y, Zhou W, et al: Inhibition of circulating miR-194-5p reverses osteoporosis through Wnt5a/β-catenin-dependent induction of osteogenic differentiation. Mol Ther Nucleic Acids. 21:814–823. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Futai M, Sun-Wada GH, Wada Y, Matsumoto N and Nakanishi-Matsui M: Vacuolar-type ATPase: A proton pump to lysosomal trafficking. Proc Jpn Acad Ser B Phys Biol Sci. 95:261–277. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Shen S, Si M, Zeng C, Liu EK, Chen Y, Vacher J, Zhao H, Mohan S and Xing W: Leucine repeat rich kinase 1 controls osteoclast activity by managing lysosomal trafficking and secretion. Biology (Basel). 12:5112023.PubMed/NCBI | |
|
Li MCM, Chow SKH, Wong RMY, Qin L and Cheung WH: The role of osteocytes-specific molecular mechanism in regulation of mechanotransduction - A systematic review. J Orthop Translat. 29:1–9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Vasiliadis ES, Evangelopoulos DS, Kaspiris A, Benetos IS, Vlachos C and Pneumaticos SG: The role of sclerostin in bone diseases. J Clin Med. 11:8062022. View Article : Google Scholar : PubMed/NCBI | |
|
Mann G, Mora S, Madu G and Adegoke OAJ: Branched-chain amino acids: Catabolism in skeletal muscle and implications for muscle and whole-body metabolism. Front Physiol. 12:7028262021. View Article : Google Scholar : PubMed/NCBI | |
|
Hatazawa Y, Tadaishi M, Nagaike Y, Morita A, Ogawa Y, Ezaki O, Takai-Igarashi T, Kitaura Y, Shimomura Y, Kamei Y and Miura S: PGC-1α-mediated branched-chain amino acid metabolism in the skeletal muscle. PLoS One. 9:e910062014. View Article : Google Scholar | |
|
Lv Z, Shi W and Zhang Q: Role of essential amino acids in age-induced bone loss. Int J Mol Sci. 23:112812022. View Article : Google Scholar : PubMed/NCBI | |
|
Takegahara N, Kim H and Choi Y: Unraveling the intricacies of osteoclast differentiation and maturation: Insight into novel therapeutic strategies for bone-destructive diseases. Exp Mol Med. 56:264–272. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Y, Cardoso FF, Parys C, Cardoso FC and Loor JJ: Branched-Chain amino acid supplementation alters the abundance of mechanistic target of rapamycin and insulin signaling proteins in subcutaneous adipose explants from lactating holstein cows. Animals (Basel). 11:27142021. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JM, Yang YS, Hong J, Chaugule S, Chun H, van der Meulen MCH, Xu R, Greenblatt MB and Shim JH: Biphasic regulation of osteoblast development via the ERK MAPK-mTOR pathway. Elife. 11:e780692022. View Article : Google Scholar : PubMed/NCBI | |
|
Carbone L, Bůžková P, Fink HA, Robbins JA, Barzilay JI, Elam RE, Isales C, Connelly MA and Mukamal KJ: Plasma levels of branched chain amino acids, incident hip fractures, and bone mineral density of the hip and spine. J Clin Endocrinol Metab. 108:e1358–e1364. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Urano T, Kuroda T, Uenishi K and Shiraki M: Serum branched-chain amino acid levels are associated with fracture risk in Japanese women. Geriatr Gerontol Int. 24:603–608. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Plotkin DL, Delcastillo K, Van Every DW, Tipton KD, Aragon AA and Schoenfeld BJ: Isolated leucine and branched-chain amino acid supplementation for enhancing muscular strength and hypertrophy: A narrative review. Int J Sport Nutr Exerc Metab. 31:292–301. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Cereda E, Pisati R, Rondanelli M and Caccialanza R: Whey protein, leucine- and vitamin-D-enriched oral nutritional supplementation for the treatment of sarcopenia. Nutrients. 14:15242022. View Article : Google Scholar : PubMed/NCBI | |
|
Ely IA, Phillips BE, Smith K, Wilkinson DJ, Piasecki M, Breen L, Larsen MS and Atherton PJ: A focus on leucine in the nutritional regulation of human skeletal muscle metabolism in ageing, exercise and unloading states. Clin Nutr. 42:1849–1865. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kitajima Y, Takahashi H, Akiyama T, Murayama K, Iwane S, Kuwashiro T, Tanaka K, Kawazoe S, Ono N, Eguchi T, et al: Supplementation with branched-chain amino acids ameliorates hypoalbuminemia, prevents sarcopenia, and reduces fat accumulation in the skeletal muscles of patients with liver cirrhosis. J Gastroenterol. 53:427–437. 2018. View Article : Google Scholar | |
|
Wang S, Guo W and Dong R: Unraveling the transcriptomic effects of leucine supplementation on muscle growth and performance in basketball athletes. PLoS One. 20:e03166032025. View Article : Google Scholar : PubMed/NCBI | |
|
Dong Y, Yuan H, Ma G and Cao H: Bone-muscle crosstalk under physiological and pathological conditions. Cell Mol Life Sci. 81:3102024. View Article : Google Scholar : PubMed/NCBI | |
|
Watanabe-Takano H, Ochi H, Chiba A, Matsuo A, Kanai Y, Fukuhara S, Ito N, Sako K, Miyazaki T, Tainaka K, et al: Mechanical load regulates bone growth via periosteal osteocrin. Cell Rep. 36:1093802021. View Article : Google Scholar : PubMed/NCBI | |
|
Klein-Nulend J, van Oers RF, Bakker AD and Bacabac RG: Nitric oxide signaling in mechanical adaptation of bone. Osteoporos Int. 25:1427–1437. 2014. | |
|
Holmes D: Bone: Irisin boosts bone mass. Nat Rev Endocrinol. 11:6892015. | |
|
Sheng R, Cao M, Song M, Wang M, Zhang Y, Shi L, Xie T, Li Y, Wang J and Rui Y: Muscle-bone crosstalk via endocrine signals and potential targets for osteosarcopenia-related fracture. J Orthop Translat. 43:36–46. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Guo Q, Duan Y, Wang W, Yang Y and Yin Y, Gong S, Han M, Li F and Yin Y: Potential nutritional healthy-aging strategy: Enhanced protein metabolism by balancing branched-chain amino acids in a finishing pig model. Food Funct. 13:6217–6232. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ikeda T, Matsunaga Y, Kanbara M, Kamono A, Masuda T, Watanabe M, Nakanishi R and Jinno T: Effect of exercise therapy combined with branched-chain amino acid supplementation on muscle strength in elderly women after total hip arthroplasty: A randomized controlled trial. Asia Pac J Clin Nutr. 28:720–726. 2019.PubMed/NCBI | |
|
Huynh H and Wan Y: mTORC1 impedes osteoclast differentiation via calcineurin and NFATc1. Commun Biol. 1:292018. View Article : Google Scholar : PubMed/NCBI | |
|
Ozaki K, Yamada T, Horie T, Ishizaki A, Hiraiwa M, Iezaki T, Park G, Fukasawa K, Kamada H, Tokumura K, et al: The L-type amino acid transporter LAT1 inhibits osteoclastogenesis and maintains bone homeostasis through the mTORC1 pathway. Sci Signal. 12:eaaw39212019. View Article : Google Scholar : PubMed/NCBI | |
|
Rivera CN, Watne RM, Wommack AJ and Vaughan RA: The effect of insulin resistance on extracellular BCAA accumulation and SLC25A44 expression in a myotube model of skeletal muscle insulin resistance. Amino Acids. 55:1701–1705. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
da Silva FMO, Pimenta AM, Juvanhol LL, Hermsdorff HHM and Bressan J: Obesity incidence according to branched-chain amino acid intake and plant-based diet index among Brazilian adults: A six-year follow-up of the CUME study. Nutrients. 17:2272025. View Article : Google Scholar : PubMed/NCBI | |
|
Holeček M: The role of skeletal muscle in the pathogenesis of altered concentrations of branched-chain amino acids (valine, leucine, and isoleucine) in liver cirrhosis, diabetes, and other diseases. Physiol Res. 70:293–305. 2021. View Article : Google Scholar | |
|
You Y, Leng S, Shi J, Yang H, Chang M, Ma Q, Zhang D, Sun H, Wang L, Gao Z, et al: Integration of bone-targeted delivery and crosstalk modulation of liver-bone axis for improved osteoporosis therapy. ACS Nano. 19:23955–23968. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zuo X, Zhao R, Wu M, Wang Y, Wang S, Tang K, Wang Y, Chen J, Yan X, Cao Y and Li T: Multi-omic profiling of sarcopenia identifies disrupted branched-chain amino acid catabolism as a causal mechanism and therapeutic target. Nat Aging. 5:419–436. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Tilg H and Moschen AR: Adipocytokines: Mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 6:772–783. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Olson OC, Quail DF and Joyce JA: Obesity and the tumor microenvironment. Science. 358:1130–1131. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Jarvis SE, Nguyen M and Malik VS: Association between adherence to plant-based dietary patterns and obesity risk: A systematic review of prospective cohort studies. Appl Physiol Nutr Metab. 47:1115–1133. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Singh L, Tyagi S, Myers D and Duque G: Good, bad, or ugly: The biological roles of bone marrow fat. Curr Osteoporos Rep. 16:130–137. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bo T and Fujii J: Primary roles of branched chain amino acids (BCAAs) and their metabolism in physiology and metabolic disorders. Molecules. 30:562024. View Article : Google Scholar | |
|
Habibi M, Shili CN, Sutton J, Goodarzi P and Pezeshki A: Dietary branched-chain amino acids modulate the dynamics of calcium absorption and reabsorption in protein-restricted pigs. J Anim Sci Biotechnol. 13:152022. View Article : Google Scholar : PubMed/NCBI | |
|
Salem A, Ben Maaoui K, Jahrami H, AlMarzooqi MA, Boukhris O, Messai B, Clark CCT, Glenn JM, Ghazzaoui HA, Bragazzi NL, et al: Attenuating muscle damage biomarkers and muscle soreness after an exercise-induced muscle damage with branched-chain amino acid (BCAA) supplementation: A systematic review and meta-analysis with meta-regression. Sports Med Open. 10:422024. View Article : Google Scholar : PubMed/NCBI | |
|
Choi BH, Hyun S and Koo SH: The role of BCAA metabolism in metabolic health and disease. Exp Mol Med. 56:1552–1559. 2024. View Article : Google Scholar : PubMed/NCBI |