key: cord-0744022-ave9ashd authors: Li, Qiangqiang; Cheng, Jack Chun‐yiu; Jiang, Qing; Lee, Wayne Yuk‐wai title: Role of sirtuins in bone biology: Potential implications for novel therapeutic strategies for osteoporosis date: 2021-01-04 journal: Aging Cell DOI: 10.1111/acel.13301 sha: 7c05e40cce6f83f3c73b0a731288d6d0d83932c5 doc_id: 744022 cord_uid: ave9ashd The decline in bone mass and bone strength and musculoskeletal problems associated with aging constitute a major challenge for affected individuals and the healthcare system globally. Sirtuins 1‐7 (SIRT1‐SIRT7) are a family of nicotinamide adenine dinucleotide‐dependent deacetylases with remarkable abilities to promote longevity and counteract age‐related diseases. Sirtuin knockout and transgenic models have provided novel insights into the function and signaling of these proteins in bone homeostasis. Studies have revealed that sirtuins play a critical role in normal skeletal development and homeostasis through their direct action on bone cells and that their dysregulation might contribute to different bone diseases. Preclinical studies have demonstrated that mice treated with sirtuin agonists show protection against age‐related, postmenopausal, and immobilization‐induced osteoporosis. These findings suggest that sirtuins could be potential targets for the modulation of the imbalance in bone remodeling and treatment of osteoporosis and other bone disorders. The aim of this review was to provide a comprehensive updated review of the current knowledge on sirtuin biology, focusing specifically on their roles in bone homeostasis and osteoporosis, and potential pharmacological interventions targeting sirtuins for the treatment of osteoporosis. Bone is a dynamic tissue that is constantly adapting its structure to functional demands through processes tightly regulated by two principal cell types: osteoblasts and osteoclasts (Harada & Rodan, 2003) . Imbalanced action of these two cell types disrupts bone formation and resorption, leading directly or indirectly to the manifestation of various bone diseases (Rodan & Martin, 2000) . Osteoporosis is a major public health concern worldwide, particularly in aging societies (Gullberg et al., 1997; Zeng et al., 2019) . Current pharmacological treatments of osteoporosis primarily aim either to reduce excessive osteoclastic bone resorption (e.g., estrogen and bisphosphonates) or to promote osteoblastic bone formation (e.g., parathyroid hormone), and to a lesser degree, to achieve both (e.g., anti-sclerostin [SOST] antibody). The primary cause of this imbalance in bone cell activity observed with aging is not yet fully understood. In addition, there is growing concern about the long-term use of these drugs due to their off-target effects (Davis et al., 2016; Kim et al., 2016; Lv et al., 2020; Silverman & Landesberg, 2009 ). There is a clear demand for continuing efforts in the research and development of safer preventative and/or therapeutic agents. Mitochondrial dysfunction (McBride et al., 2006; Trifunovic & Larsson, 2008; Trifunovic et al., 2004; Varanasi et al., 1999) , oxidative stress (Balaban et al., 2005; Cadenas & Davies, 2000) , and inflammation (El Assar et al., 2013; Woods et al., 2012) are found to be closely related with the impairment of cellular homeostasis and contribute to the progression of age-related diseases, including osteoporosis. Therefore, targeting these primary aging processes represents a new class of treatment strategies for multiple aging tissues, including bone. Mammalian sirtuins play important roles in longevity, aging-associated diseases, and response to stresses (Bonkowski & Sinclair, 2016; Donmez & Guarente, 2010; Haigis & Sinclair, 2010; Imai & Guarente, 2014; Wątroba et al., 2017) . Increased sirtuin activity is associated with the delayed onset of age-related diseases, including cancer, cardiovascular disease, and diabetes (Banks et al., 2008; Hubbard & Sinclair, 2014; Kanfi et al., 2010; Kugel et al., 2016; Pfluger et al., 2008) , and prolonged longevity in some circumstances (Kanfi et al., 2012; Mercken et al., 2014; Mitchell et al., 2014; Satoh et al., 2013) . These beneficial effects might be attributable to the action of sirtuins in mitochondrial biogenesis, increased resistance against oxidative stress, and anti-inflammatory effects (Donmez & Guarente, 2010; Finkel et al., 2009; Haigis & Sinclair, 2010; Hirschey et al., 2010; Kincaid & Bossy-Wetzel, 2013; Rardin et al., 2013) . It has been postulated that manipulation of sirtuin expression and/or activity could represent a new therapeutic approach for the prevention and treatment of osteoporosis. In this review paper, we present a comprehensive updated review of the current knowledge on sirtuin biology, focusing specifically on their roles in bone homeostasis and osteoporosis, and the potential pharmacological interventions that could target sirtuins for the treatment of osteoporosis. Osteoporosis is the most common bone remodeling disease, and the resulting increased risk of fragility fractures is of great concern (Pouresmaeili et al., 2018) . Notably, the risk of fracture doubles with every 10% of bone mass lost (Rodan & Martin, 2000) . The number of people who experience hip fracture in the United States has been estimated to reach over 6.26 million by 2050, resulting in dramatically high morbidity and healthcare costs (Gullberg et al., 1997) . In China, there are currently over 60 million individuals diagnosed with osteoporosis, and the prevalence is estimated to be 6.49% and 29.13% for men and women aged 50 years or older, respectively (Zeng et al., 2019) . Considering the huge social and economic burden associated with osteoporosis, there is an urgent need for a better understanding of the molecular mechanism underlying osteoporosis which might provide a scientific basis for the development of more effective therapies. Recent clinical and preclinical animal studies have demonstrated an association between increased mitochondrial damage and osteoporotic bone loss (Trifunovic & Larsson, 2008; Trifunovic et al., 2004; Varanasi et al., 1999) . Mice lacking HTRA2/OMI, an mitochondrial adenosine triphosphate (ATP)-independent serine protease, exhibited elevated mtDNA deletions and severe osteoporosis . Impaired ATP production, caused by a dysfunctional mitochondrial transcription factor A (Tfam) gene, led to increased bone resorption (Miyazaki et al., 2012) . Mitophagy and the unfolded protein response (UPR mt ) are important for mitochondrial homeostasis (Fang et al., 2014; Mouchiroud et al., 2013b) , whereas their disruption leads to severe bone loss, which is closely associated with impaired mitochondrial function (Wang et al., 2020; Zainabadi, Liu, Caldwell, et al., 2017) . Furthermore, in vitro cellular and in vivo animal studies have demonstrated a link between mitochondria-derived reactive oxygen species (ROS) and osteoporosis (Manolagas, 2010; Treiber et al., 2011; Yang et al., 2014) . The skeletal changes in aging mice were accompanied by a progressive increase in ROS levels in the bone tissues (Almeida et al., 2007) , which could adversely influence the survival and differentiation of osteoblasts and osteocytes from their corresponding progenitor cells. Mice with homozygous superoxide dismutase 2 (SOD2) deficiency in connective tissue (established using Col1α2-Cre) exhibited decreased bone mineral density (BMD) associated with increased ROS levels (Treiber et al., 2011) . Specific deletion of SOD2 in osteocytes caused remarkable bone loss in an age-dependent manner, which was associated with the decreased number of osteocytes and disorganization of osteocyte canalicular networks resulting from increased ROS levels (Kobayashi et al., 2015) . High levels of hydrogen peroxide (H 2 O 2 ) in osteoblastic cells resulted in apoptosis and initiated osteoporosis by impairing osteoblast formation (Treiber et al., 2011; Yang et al., 2014) . Conversely, accumulation of ROS induced osteoclast proliferation and facilitated osteoclast differentiation (Baek et al., 2010; Bartell et al., 2014; Garrett et al., 1990; Treiber et al., 2011; Yang et al., 2014) . Loss of forkhead box O (FoxO) function in osteoclasts in mice increased osteoclast numbers and resulted in bone loss due to accumulation of intracellular H 2 O 2 . Attenuation of H 2 O 2 generation in cells of the osteoclast lineage (using the lysozyme M promoter) abolished the loss of cortical bone following ovariectomy (OVX) by decreasing osteoclast numbers (Ucer et al., 2017) . Collectively, factors modulating mitochondrial function and antioxidant defense could be potential targets for interventional treatment or prevention of osteoporosis. Various intra-and extracellular stresses, such as DNA damage, oncogenic insults, reactive metabolites, and proteotoxic stress, lead to cellular senescence Swanson et al., 2013; Tchkonia et al., 2013; Zhu et al., 2014) , in which cells stop dividing, undergo distinct genetic and phenotypic changes, and develop a senescence-associated secretory phenotype (SASP; Blasco et al., 2013; Campisi, 2013) . SASP is characterized by the secretion of pro-inflammatory cytokines, chemokines, and extracellular matrixdegrading proteins, which have deleterious paracrine and systemic effects (Acosta et al., 2013; Coppé et al., 2010; Nelson et al., 2012; Xu, Palmer, et al., 2015) . By secreting damaging factors to neighboring cells, the SASP induces cellular senescence in normal cells adjacent to the senescent cells via the bystander effect (Nelson et al., 2012) . Reducing the number of senescent cells by either genetic (INK-ATTAC transgenic mice) or pharmacological (long-term senolytic treatment) clearance of p16 Ink4a -expressing senescent cells has been shown to extend the life span and prevent the development of multiple aging-related comorbidities in both prematurely and naturally aged mice (Baker et al., 2011; Roos et al., 2016; Xu, Palmer, et al., 2015) . A recent study comparing young (6-month-old) and aged (24-month-old) mice demonstrated that osteocytes and myeloid cells were the two main senescent cells in the bone microenvironment (Farr et al., 2016) . Increased osteocyte senescence was further confirmed by another study that showed higher expression of the senescence markers γH2AX and p16 Ink4a , and several SASP markers in osteocytes in 21-month-old in comparison with their expression levels in 7-month-old mice (Piemontese et al., 2017) . A subsequent study from the same group demonstrated similar findings in osteoprogenitors. Specifically, a decreasing number of osterix (Osx)-expressing osteoprogenitor cells were observed with increasing age in mice and were associated with increased expression of senescence markers (Kim, Chang, et al., 2017) . Furthermore, bone marrow stromal cells from old mice also exhibited elevated expression of SASP genes (Kim, Chang, et al., 2017; Sui et al., 2016) . In addition to these findings on cellular senescence in age-related bone loss, increased cellular senescence in bone cells was also associated with glucocorticoid-induced (Leclerc et al., 2004; Li et al., 2012) and unloading-induced (Okazaki et al., 2004; Sakai et al., 2002) bone loss. The causal relationship between cellular senescence and age-related bone loss was evidenced by the higher bone mass and strength observed in aged mice after the elimination of senescent cells in bone tissue by activating transduced suicide genes or administrating senolytics or a JAK inhibitor (Farr et al., 2017; Kim, Chang, et al., 2017) . It has been proposed that SASP-associated factor secretion from the senescent osteoblasts and osteocytes could lead to increased bone resorption and reduced bone formation. Thus, targeting senescent osteocytes represents a novel treatment for age-related bone loss. Furthermore, cell cycle arrest is part of cellular senescence, which limits osteoblast differentiation of skeletal progenitor cells and contributes to age-related bone loss . Future in-depth studies are needed to investigate the mechanisms by which cellular senescence could impact bone remodeling. In view of current evidence demonstrating beneficial effects, eliminating senescent cells in the bone microenvironment represents a novel strategy that may potentially be used for the prevention and treatment of osteoporosis. Under the influence of lifelong exposure to chronic antigenic load and oxidative stress during aging, a variety of cytokines, including interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), and interleukin 1 (IL-1), are elevated and play direct roles in the pathogenesis of age-related diseases, including atherosclerosis, Alzheimer's disease and diabetes (Bruunsgaard, 2002; Ferrucci & Fabbri, 2018; Sarkar & Fisher, 2006; Yuan et al., 2019) . Recent emerging evidence suggests that osteoporosis and other age-related disorders could, to certain extent, be considered as inflammatory diseases (Arron & Choi, 2000; Lorenzo, 2000) . Clinical studies have reported an association between an increased risk of developing osteoporosis and inflammatory conditions, such as rheumatoid arthritis, ankylosing spondylitis, and inflammatory bowel disease (Bultink et al., 2005; Haugeberg et al., 2004; Mikuls et al., 2005; Mitra et al., 2000; Moschen et al., 2005) . Inflammatory cytokines, such as TNF-α, IL-1, and IL-6, are elevated and play critical roles in these conditions (Ishihara & Hirano, 2002; Manolagas & Jilka, 1995; Moschen et al., 2005) . Specifically, the aforementioned cytokines promote osteoclast differentiation and activation, which has been linked to accelerated bone loss in various bone disorders including postmenopausal osteoporosis, Paget's disease, and idiopathic osteoporosis (Kim et al., 2009; Manolagas & Jilka, 1995; Moffett et al., 2005; Pacifici et al., 1989; Wei et al., 2005; Yun & Lee, 2004) . In women, the most common cause of postmenopausal osteoporosis is estrogen depletion, which results in elevated levels of pro-inflammatory and pro-osteoclastic cytokines (Ershler et al., 1997; Liu et al., 2005; Pfeilschifter et al., 2002; Scheidt-Nave et al., 2001) . These findings support the observation that inflammation could exert significant influence on bone turnover and contribute to the development of osteoporosis. Regulation of rRNA transcription, cell cycle regulation, and ribosome biogenesis Deacetylase p53, tumor protein p53 ; FOXO1/3/4, forkhead box O-1/3/4; p300, histone acetyltransferase p300; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PGC1α, peroxisome proliferatoractivated receptor alpha; LKB1, liver kinase B1; PPARγ, peroxisome proliferator-activated receptor gamma; ATG5/7/8, autophagy-related gene 5/7/8; MyoD, myogenic differentiation 1; MEF2, myocyte enhancer factor 2; XPA, xeroderma pigmentosum, complementation group A; XPC, xeroderma pigmentosum, complementation group C; NBS1, Nijmegen breakage syndrome 1; HSF1, heat shock transcription factor 1; BCL6, B-cell lymphoma 6 protein; SMAD3/7, SMAD family member 3/7; MLH1, MutL homolog 1; PMS2, PMS1 homolog 2; HIF, hypoxia-inducible factor; PTEN, phosphatase and tensin homolog; AR, androgen receptor; DNMT1, DNA methyltransferase 1; AceCS1, Acyl-CoA synthetase short-chain family member 1; eNOS, endothelial nitric oxide synthase; ING3, inhibitor of growth family member 3; UPC2, ultraperformance convergence chromatography; ACS1, acetyl-CoA synthetase 1; CRTC2, CREB-regulated transcription coactivator 2; PGAM-1, phosphoglycerate mutase-1; SREBP, sterol regulatory element-binding protein; LXR, liver X receptor; FXR, farnesoid X receptor; HDAC6, histone deacetylase 6; HOXA10, homeobox protein A10; CDK1, cyclin-dependent kinase 1; LCAD, long-chain-specific acyl coenzyme A dehydrogenase; ACS2, acetyl-CoA synthetase 2; IDH2, isocitrate dehydrogenase 2; Ku70, lupus Ku autoantigen protein p70; MnSOD, mitochondrial manganese superoxide dismutase; MRPL10, mitochondrial ribosomal protein L10; HMG-CoA2, 3-hydroxy-3-methylglutaryl CoA synthase 2; SDH, succinate dehydrogenase; NADH, nicotinamide adenine dinucleotide; OTC, ornithine transcarbamoylase; GDH, glutamate dehydrogenase; MCD, malonyl CoA decarboxylase; PDH, pyruvate dehydrogenase; ANT2, adenine translocator 2; ANT3, adenine translocator 3; IDE, insulin-degrading enzyme; Cyt c, cytochrome c; CPS1, carbamoyl phosphate synthetase 1; UOX, urate oxidase; TNF-α, tumor necrosis factor α; PAPR1, poly (adenosine diphosphate-ribose) polymerase-1; HIF1α, hypoxia-induced factor 1; NPM1, nucleophosmin 1; KAP1, the Krüppel associated box (KRAB)-associated protein-1; GCN5, general control non-repressed protein 5 (an acetyltransferase); PGK1, phosphoglycerate kinase 1; CDK9, cyclin-dependent kinase 9; DDB1, damage specific DNA binding protein 1; SMAD4, mothers against decapentaplegic homolog 4. The first member of the sirtuin family, yeast Sir2 (silent information regulator 2), was isolated in a screening for silencing factors in budding yeast thirty years ago (Rine et al., 1979) . Subsequent studies revealed that SIR2-like genes, also known as sirtuins, play a key role in prolonging the life span of lower organisms (Kaeberlein et al., 1999; Kennedy et al., 1995) . The important discovery that life span extension by caloric restriction is abolished in sirtuin-deficient yeast has inspired multiple studies on mammalian sirtuins (Guarente & Picard, 2005; Boily et al., 2008) . To date, seven mammalian homologs (SIRT1-SIRT7) of nicotinamide adenine dinucleotide (NAD + )-dependent lysine deacetylases have been identified and found to be important in the promotion of DNA repair, antioxidant defense, mitochondrial biogenesis, and anti-inflammatory effects (Donmez & Guarente, 2010; Finkel et al., 2009; Haigis & Sinclair, 2010) . These sirtuins differ from each other in tissue distribution, subcellular localization, enzymatic activity, and target proteins (Table 1) Among the seven sirtuins, SIRT1 is the most studied and plays critical role in normal skeletal development and homeostasis (Zainabadi, 2019) . This review attempts to provide more updated information regarding the role of SIRT1 in bone biology and osteoporosis development. Pioneering work on the role of SIRT1 in bone biology was conducted in SIRT1 global knockout (KO) mice (Cheng et al., 2003; Lemieux et al., 2005; McBurney et al., 2003) . Compared with wild-type control mice, SIRT1 KO embryos and newborn pups were smaller at birth and exhibited higher rates of perinatal lethality and notable developmental defects of the retina and heart ( Figure 1 ). These mice also had craniofacial abnormalities, including defects in the development and closure of craniofacial sutures, abnormal palate architecture, and instances of exencephaly (Cheng et al., 2003; McBurney et al., 2003) . Furthermore, SIRT1 KO pups exhibited delayed mineralization of the skull, vertebrae, and digits (Lemieux et al., 2005) . Likewise, deletion of SIRT1 in adult mice showed a significant reduction in both trabecular and cortical bone irrespective of sex Zainabadi, Liu, Caldwell, et al., 2017) . These results suggest an important role of SIRT1 in bone development and remodeling. However, it is difficult to interpret these findings separately from the frequently associated gross developmental abnormalities, including smaller size, sterility, and high rates of postnatal lethality (Cheng et al., 2003; McBurney et al., 2003) . Additional observations of adult heterozygous KOs showed a significant reduction in both trabecular and cortical bone mass in long bones (Cohen-Kfir et al., 2011) without developmental abnormalities, thus supporting the hypothesis that SIRT1 plays a role in bone remodeling. Interestingly, the phenotypic changes appear to be sex-and age-specific, as in- using the lysozyme M promoter showed lower trabecular bone mass (Zainabadi, Liu, Caldwell, et al., 2017) . However, double deletion of SIRT1 in both osteoblasts and osteoclasts did not result in a more severe bone loss phenotype (Zainabadi, Liu, Caldwell, et al., 2017) . Mechanistic studies revealed that the bone loss phenotype of ObcKOs was associated with a lower osteoblast number and reduced bone formation rate (BFR), while increased osteoclast number was observed in OccKOs. These findings support the hypothesis that SIRT1 can exert a direct effect on osteoblast and osteoclast. Furthermore, deletion of SIRT1 in osteoprogenitors using the Osx promoter resulted in lower cortical bone thickness at the endocortical surface as a result of decreased bone formation, but had no effect on trabecular bone mass . Similar phenotypic changes were observed in mice with the deletion of SIRT1 in earlier osteoblast precursors using the Prx1 promoter (Simic et al., 2013) . These results, along with the finding that cortical bone was unaffected in mice lacking SIRT1 in mature osteoblasts (Edwards et al., 2013) , suggest that SIRT1 using the collagen II promoter resulted in growth retardation and shorter bones, which was associated with decreased chondrocyte proliferation and hypertrophy (Jin et al., 2019) . In addition, SIRT1 has been shown to promote cartilage-specific gene expression (Dvir-Ginzberg et al., 2008) and protect chondrocyte against radiation-induced senescence (Hong et al., 2010) . These findings were consistent with reduced levels of SIRT1 measured in human osteoarthritis (OA) cartilage (Fujita et al., 2011; Takayama et al., 2009) , suggesting a potential protective role of SIRT1 in chondrocytes, which was further evidenced by the accelerated development of OA in mice lacking SIRT1 in chondrocyte (Matsuzaki et al., 2014) . However, a recent finding demonstrated an increased ratio of proliferative-to-hypertrophic zone following SIRT1 deletion in chondrocyte, suggesting SIRT1 inhibits chondrocyte proliferation (Shtaif et al., 2020) . Possible reason for the discrepancy could be due to the differences in the genetic models, which requires further investigation. In addition to its role in chondrogenesis, the absence of SIRT1 in chondrocytes also resulted in decreased trabecular and cortical bone mineralization, which was attributed to the disorganized epiphyseal growth plate in chondrocyte-specific KO mice (Shtaif et al., 2020) . In addition to the KO models, SIRT1 overexpression mouse models have also been developed to study the impact of SIRT1 activation on bone homeostasis (Herranz et al., 2010; Sun et al., 2018; Wang et al., 2019) . Encouragingly, these transgenic mice showed 40%-50% higher bone mass than that of control mice at 2.5 years of age (Herranz et al., 2010) , as well as improved healthy aging. More recently, overexpression of SIRT1 in cells of the mesenchymal lineage (using the Prx1 promoter) showed increased bone volume in both young mice (1-month-old) and aged mice (18-month-old), which was associated with increased osteoblast and decreased osteoclast numbers (Sun et al., 2018; Wang et al., 2019) . These effects were achieved by restoring the redox balance in MSCs. These preclinical findings suggest that SIRT1 could be a potential target for the development of novel anti-osteoporotic therapy. SIRT2 KO mice developed normally at a young age but exhibited tumorigenesis in multiple tissues at an advanced age, suggesting an essential role of SIRT2 in maintaining genetic stability and repressing tumor formation . Among all reported SIRT2 KO studies (Beirowski et al., 2011; Jing et al., 2019; Kim et al., 2011) , one related to skeleton changes was identified (Jing et al., 2019) and is included in the present review. Compared with the wild-type control group, rats with SIRT2 deletion (SIRT2 KO) exhibited a higher total bone volume fraction, trabecular bone mineral density, and trabecular number at the age of 36 weeks, which was not apparent at the age of 12 weeks. In an in vitro study, it was demonstrated that inhibiting SIRT2 expression using AGK2 could suppress osteoclastogenic differentiation (Jing et al., 2019) . Further investigation is required to delineate how SIRT2 regulates osteoclast differentiation and activity, and its effect on age-related bone loss. SIRT3, a mitochondrial NAD + -dependent protein deacetylase (Ansari et al., 2017; Lombard et al., 2007) , was found to be associated with long life span in humans and has, thus, attracted public and research interest (Bellizzi et al., 2005; Rose et al., 2003) . An initial study showed that SIRT3 KO mice appeared to be structurally normal, without significant changes in body composition and BMD at a young age (Lombard et al., 2007) . However, aging-related health problems such as metabolic syndrome, cancer, and cardiovascular and neurodegenerative diseases were observed earlier in , which was associated with impaired bone formation. The effects of SIRT6 on bone resorption remain controversial, with only one report of impaired osteoclast function (Sugatani et al., 2015) . In contrast, most other studies have reported increased osteoclastogenesis reported (Zhang et al., 2016a . Interpretation of these findings is difficult due to the extremely small size of the tested animals and the overall poor health condition of the SIRT6 KO mice. To better understand the exact molecular role of SIRT6 in bone homeostasis, several osteoblast lineage-and osteoclast lineage-specific SIRT6 KO mice have been developed. Specifically, osteoblast lineage-specific SIRT6 KO mice established using the osteocalcin promoter exhibited osteopenia, which was attributed to a paracrine activation of osteoclastogenesis due to decreased osteoprotegerin (OPG) levels without changes in osteoblast function (Kim et al., 2020) . Targeted deletion of SIRT6 in hematopoietic cells (using the Mx1 promoter), including osteoclast precursors, resulted in increased bone volume, which could be due to the decreased number of osteoclasts . In contrast, myeloid SIRT6 deletion (using the lysozyme M promoter) resulted in decreased trabecular bone mass in aged mice (20-month-old). This age-related change in phenotype was associated with a twofold increase in the number of mature osteoclasts in SIRT6 KO mice (Moon et al., 2019) . These results, along with the controversial findings on the effects of SIRT6 on osteoclasts, suggest that a more in-depth elucidation of the role of SIRT6 in skeletal homeostasis is required, such as in cKO and transgenic animal models. collagen promoter) showed decreased trabecular bone mass and cortical bone thickness (Fukuda et al., 2018) . Importantly, the phenotype of obcKOs was found to be associated with lower in vivo osteoblast numbers and reduced BFR, with no effect on osteoclastogenesis. The stimulatory effects of SIRT7 on osteoblasts might be due to its positive regulation of the transactivation activity of Osx through deacetylation of lysine 368 in its C-terminal region. However, there is no available data on the influence of SIRT7 on osteoclast function, which requires further investigation. The (Shakibaei et al., 2012; Tseng et al., 2011; , and repressing peroxisome proliferator-activated receptor γ (PPARγ; Bäckesjö et al., 2006; Picard et al., 2004; Figure 2b ). In addition, SIRT1 has been shown to promote osteoblast activity by stimulating Wnt signaling through deacetylation effect directly (Simic et al., 2013) or indirect deacetylation of FoxOs, which prevents the association of FoxO with β-catenin, thus resulting in increased expression of β-catenin . SIRT1 may also promote bone formation by decreasing the expression of SOST (Artsi et al., 2014; Cohen-Kfir et al., 2011; Stegen et al., 2018) , an inhibitor of bone formation, by modifying H3 K9 acetylation at the Sost promoter, silencing SOST expression. Additionally, SIRT1 activates PGC1α transcriptional activity to induce mitochondrial biogenesis and the induction of antioxidative enzymes, which can protect bone cells and inhibit the generation of mitochondrial ROS (Cantó et al., 2009; He et al., 2010; Yao et al., 2018) . In addition to its stimulatory actions on osteogenic differentiation from bone mesenchymal stromal cells (BMSCs), SIRT1 has also been shown to play a role in mature osteoblasts (Figure 2c ). Upregulation of SIRT1 inhibits H 2 O 2 -induced osteoblast apoptosis via the FoxO1/β-catenin pathway (Yao et al., 2018) . Similarly, SIRT1 has been shown to suppress osteoblast apoptosis by inhibiting both the p53-p21 and NF-κB signaling pathways (Gu et al., 2019; Huang et al., 2012) . Furthermore, SIRT1 has been shown to repress osteoclast differentiation through negative regulation of NF-κB and positive regulation of FoxO transcription factors (Edwards et al., 2013; Kim et al., 2015 ; Figure 3a ). The roles of sirtuins in osteocytes are less studied to date. A recent study demonstrated that SIRT1-dependent deacetylation of the Sost promoter resulted in decreased sclerostin expression and enhanced WNT/β-catenin signaling, leading to increased bone mass in mice (Stegen et al., 2018 ; Figure 3b ). To date, the roles of other sirtuins in bone cells in vitro have not been extensively studied. Inhibition of SIRT2 with its specific inhibitor, AGK2, suppressed the differentiation of BMMs into osteoclasts by reducing the expression of c-Fos and nuclear factor of F I G U R E 2 Sirtuins regulate BMSCs, osteogenic differentiation, and osteoblast function through multiple downstream targets. (a) SIRT1 maintains BMSC self-renewal and stemness through its direct regulation of SOX2. (b) SIRT1 regulates osteogenic differentiation of BMSCs by directly or indirectly interacting with RUNX2, β-catenin, SOD2, PPARγ, FOXOs, PGC1α, and SOST, thus modulating the expression of their target genes. SIRT3 decreases ROS production by stimulating SOD2, and SIRT3 also enhances cellular respiration by increasing the activities of complex I, complex II, complex III, and IDH2. (c) SIRT1 regulates the function of osteoblast by directly or indirectly regulating β-catenin, FOXO3a, and the p53-p21 and NF-κB signaling pathways, modulating the expression of their targets. SIRT3 decreases ROS production by stimulating SOD2. SIRT6 interacts with RUNX2 and OSTERIX via deacetylating H3 K9 in their promoters and increasing the expression of their target genes. SIRT7 promotes the transcriptional activity of OSTERIX by deacetylating its promoters and increasing expression of its target genes. BMSCs, bone mesenchymal stromal cells; SOD2, sex-determining region Y-box 2; RUNX2, regulating runtrelated transcription factor 2; SOD2, superoxide dismutase 2; PPARγ, peroxisome proliferator-activated receptor γ; FOXOs, forkhead box O transcription factors; PGC1α, peroxisome proliferator-activated receptor γ coactivator 1α; SOST, sclerostin; ROS, reactive oxygen species; IDH2, isocitrate dehydrogenase 2; FOXO3a, forkhead transcription factor 3a; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; H3 K9, histone H3 at lysine 9; MPP, matrix processing peptidase; Ac, acetylation; SDH, succinate dehydrogenase; TCA, tricarboxylic acid cycle in BMMs enhanced osteoclast differentiation through modulation of ROS, which stimulated the differentiation and activation of osteoclasts . These findings are in contrast to a recent finding that demonstrated that SIRT3 positively regulated osteoclastogenesis by activating the mammalian target of rapamycin (mTOR) pathway (Ho et al., 2017) . In addition to the different experimental conditions and materials used, such as BMMs of different origins. Impaired ATP production following SIRT3 F I G U R E 3 Sirtuins regulate osteoclast and osteocyte function through multiple downstream targets. (a) SIRT1 inhibits the activity of the NF-κB protein complex through the deacetylation of lysine 310 on the p65 subunit and thus decreases the expression of its target genes. SIRT3 decreases ROS production by stimulating SOD2 in mitochondria. Excessive ROS levels stimulate autophagy and promote the release of proteolytic enzymes (e.g., CTSK, MMP9) from lysosomes to resorb bone. Furthermore, SIRT3 regulates ATP generation, which is required for the generation of hydrogen ions released through vacuolar ATPase proton pumps to create an acidic environment in the resorptive pit. SIRT6 deacetylates the ERα protein to prevent its proteasomal degradation and promotes transcription of FasL in preosteoclasts, resulting in osteoclast apoptosis. (b) SIRT1-dependent deacetylation of the Sost promoter results in decreased sclerostin expression and enhanced WNT/β-catenin signaling. NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; ROS, reactive oxygen species; SOD2, superoxide dismutase 2; CTSK, cathepsin K; MMP9, matrix metalloproteinase-9; ATP, adenosine triphosphate; ADP, adenosine diphosphate; ERα, estrogen receptor alpha; FasL, fas ligand deficiency could also explain the above discrepancy due to the high energy demand of osteoclastic bone resorption (Ishii et al., 2009 ). In addition, autophagy is involved in osteoclastic bone resorption, which is regulated by ROS levels (Scherz-Shouval & Elazar, 2011; Figure 3a ). Deletion of SIRT6 in BMSCs resulted in impaired osteogenesis (Sun et al., 2014; Zhang, Cui, et al., 2016) , whereas overexpression of SIRT6 impaired osteoblast differentiation of human MSCs (Xiao et al., 2019) . The exact mechanism by which SIRT6 affects osteogenesis requires further investigation. SIRT6 has been shown to promote osteoblast activity through direct interaction with RUNX2 and Osx via deacetylating histone H3 at lysine 9 (H3 K9) in their promoters (Sugatani et al., 2015) . Furthermore, SIRT6 has also been shown to regulate osteoclastogenesis. The absence of SIRT6 impaired osteoclastogenesis associated with increased OPG expression Sugatani et al., 2015) , whereas SIRT6 deficiency was reported to induce activation of osteoclasts by inhibiting transcription of Fas ligand (FasL) in preosteoclasts, resulting in an increased number of osteoclasts (Moon et al., 2019) . Details of the regulation mechanisms need to be further clarified. A more recent study showed that SIRT7 knockdown accelerated osteogenesis of human BMSCs, at least in part via the Wnt/β-catenin signaling pathway . In this regard, it is noteworthy that SIRT7 acts on multiple cell types to regulate bone metabolism. (Hubbard & Sinclair, 2014) , 7 (Pillai et al., 2015 ), 8 (Rahnasto-Rilla et al., 2018 ), 9 (Pearson et al., 2008 , 10 (Feng et al., 2014) , 11 (H. Zhang, Ryu, et al., 2016) , 12 (Fang et al., 2016) , 13 (Zainabadi, Liu, Caldwell, et al., 2017) , 14 (Stegen et al., 2018) , 15 , 16 (Artsi et al., 2014) , and 17 (Cheng et al., 2018) molecules. The activation of sirtuins either by genetic or by pharmacological means extends life span and promotes the health of a wide variety of organisms (Kaeberlein et al., 1999; Kanfi et al., 2012; Mercken et al., 2014; Mitchell et al., 2014; Satoh et al., 2013) , thus demonstrating the feasibility of developing drugs that target sirtuins in the treatment of osteoporosis. Resveratrol, the first generation of SIRT1 activator discovered in 2003, is a polyphenol found in nuts, grapes, and other plant sources, and can increase SIRT1 protein expression in a dose-dependent manner and extend the life span of yeast (Howitz et al., 2003) . Cumulating evidence has demonstrated the protective effect of resveratrol on osteoporosis . Mice treated with resveratrol for over 18 months showed moderate improvements in both trabecular and cortical bone mass, indicating protective effect of SIRT1 activation on agingrelated bone loss Figure 4a ). Furthermore, resveratrol treatment protected against bone loss in OVX models of postmenopausal osteoporosis (Feng et al., 2014) . In addition to the osteoporosis model, resveratrol administration for 10 weeks also increased bone mass and osteoblast number in young mice (12-week-old; Zhao et al., 2018) . However, resveratrol may be able to activate SIRT3 (Chen et al., 2015) and SIRT5 (Gertz et al., 2012) as well as other non-sirtuin targets, including AMPK (Fulco et al., 2008) , F1-ATPase (Gledhill et al., 2007) , and PARp1 and Thr-tRNA (Sajish & Schimmel, 2015) . To avoid this, more specific and selective SIRT1 activators with a greater substrate-binding affinity for SIRT1 have been developed. SRT1720, SRT2104, and SRT3025 are representative sirtuin-activating compounds (STACs) discovered in the past two decades. SRT1720 was found to be beneficial for preventing bone loss. For instance, femoral trabecular bone mass in aged mice (12-month-old) increased by 50% after five months' treatment of SRT1720 (Zainabadi, Liu, Caldwell, et al., 2017) . Similarly, administration of SRT1720 attenuated bone loss in OVX models (Zainabadi, Liu, Caldwell, et al., 2017) . These benefits could be attributed to the stimulation of bone formation, inhibition of resorption, and reduction in sclerostin levels (Stegen et al., 2018) . Among the various STACs, SRT2104 not only extended the life span in mice, but also enhanced BMD by more than 65% compared with that of control mice, which might be a result of higher mitochondrial content and decreased inflammation . SRT3025 was found to significantly restore bone mass and structure in OVX mice by inhibiting the expression of sclerostin (Artsi et al., 2014) . In addition, drugs targeting SIRT1 may also have the potential to treat other bone disorders. For example, activation of SIRT1 may protect against bone loss caused by the use of glucocorticoids and thiazolidinedione, both of which are associated with impaired osteoblast differentiation and, at least in part, due to the diversion of MSCs toward the adipocyte lineage (Briot & Roux, 2015; Hou et al., 2019; Schwartz, 2008) . Similarly, the SIRT1 agonist SRT2104 may help alleviate immobilization-induced osteoporosis , a common indication in the aging population. Finally, modulation of SIRT1 expression may prove effective for combatting Paget's disease, which is the second most common bone disorder after osteoporosis, both of which are associated with defects in bone remodeling (Feng & McDonald, 2011) . These findings provide compelling evidence suggesting that SIRT1 may serve as a potential therapeutic target for combating osteoporosis and other bone disorders. Honokiol is a natural biphenolic compound derived from the bark of magnolia trees that can bind to SIRT3 and increase SIRT3 expression and activity (Pillai et al., 2015 (Pillai et al., , 2017 . Honokiol has been reported to block agonist-induced and pressure overload-mediated heart failure (Pillai et al., 2015) as well as the associated mitochondrial damage and cell death (Pillai et al., 2017) , and was accompanied by upregulation of SIRT3 expression and activity. At the time of writing, there have been no studies exploring the effect of honokiol in osteoporosis models. In addition, the aforementioned protective effect of resveratrol on bone might be mediated, at least in part, by SIRT3 as the protective effects of resveratrol were blunted by deletion of SIRT3 (Chen et al., 2015) . Therefore, STACs should be used in combination with sirtuin transgenic mice in future mechanistic studies to better understand the roles of each sirtuin in bone. In 2016, You and colleagues reported the first synthetic SIRT6 activator through screening of pyrrolo[1,2-a]quinoxaline derivatives (You et al., 2017) . Later, cyanidin was found to be a more potent SIRT6 activator and shown to increase both the expression and activity of SIRT6 (Rahnasto-Rilla et al., 2018; You et al., 2019) . Cyanidin could protect cells against oxidative stress, thus reducing the risk of age-related diseases (Kumar and Pandey, 2013; Smeriglio et al., 2016) . A recent animal study demonstrated that cyanidin treatment protected against bone loss in OVX mice by inhibiting osteoclast formation and bone resorption (Cheng et al., 2018) . More studies on cyanidin in other osteoporosis models are needed to test its therapeutic effect on bone loss prevention and potentially lead to clinical trials in the future. More recently, using an activity-based screening platform, a variety of compounds were found to be able to stimulate SIRT6-mediated deacetylation against the H3 acetyl-lysine 9 peptide. It is interesting to note that the majority of these activators consist of a terminal negative charge and linear aliphatic chain. Using further simulation analysis, the same group developed a more potent SIRT6 activator, CL5D (Klein et al., 2020) . All these findings provide an important foundation for understanding the physiological function of SIRT6 in vivo and to investigate its potential in osteoporosis treatment. NAD + is an essential sirtuin substrate, and the abundance of NAD + regulates the enzymatic activity of sirtuins (Imai & Guarente, 2014; Sauve et al., 2006) . Restoration of NAD + improved both the life span and health span in animal models of the Werner syndrome, which is accompanied by severe osteoporosis Oshima et al., 2017) . NAD + boosters serve as another class of STACs that restore NAD + levels and potentially activate all seven sirtuins (Bonkowski & Sinclair, 2016) . Nicotinamide mononucleotide (NMN), a key NAD + intermediate, has been shown to enhance NAD + biosynthesis and ameliorate various pathologies in mouse models (Imai & Guarente, 2014; Mills et al., 2016) . Mills et al. (2016) showed that NMN could provide modest protection against osteoporosis in mice. Nicotinamide riboside (NR), another natural precursor of NAD + , has been shown to extend the life span in both wild-type mice (Zhang, Ryu, et al., 2016) and progeria animal models (Fang et al., 2014 (Fang et al., , 2016 . In addition, NR also prevents high-fat diet-induced glucose dysregulation (Cantó et al., 2012) , noise-induced hearing loss (Brown et al., 2014) , cardiac injury (Xu, Barrientos, et al., 2015) , and stem cell-niche depletion (Zhang, Ryu, et al., 2016) . Unfortunately, bone phenotypes were not included in the above reports. Future studies are warranted to investigate the effect of NR on bone mass in animal models of osteoporosis. Another question to be answered is whether NAD + precursors exert their protective effects by activating all or some sirtuins. Currently, more than 100 ongoing clinical trials are evaluating the safety and physiological activity of STACs for treating human diseases such as cardiovascular diseases, type 2 diabetes, and inflammation (Baksi et al., 2014; Bonkowski & Sinclair, 2016; Figure 4b) . Some of these studies have shown the benefits of sirtuins on the cardiovascular system and metabolic diseases, while others reported little or no effects. Resveratrol is the most studied STAC in clinical trials, with NR playing a promising role (Figure 4c ). To the best of our knowledge, none of these STACs have been successfully translated into clinical practice for various reasons (those under clinical trial are not accounted for). However, resveratrol, due to its low bioavailability and potency, has demonstrated variable efficacy in humans (Tomé-Carneiro et al., 2013) . Synthetic STACs, such as SRT1720, failed to enter clinical trials due to limited target specificity (Dai et al., 2018; Huber et al., 2010; Nguyen et al., 2013) . On the other hand, SRT2104, which mimics the effects of caloric restriction and extends the life span of male mice , has entered phase II trials with few side effects Venkatasubramanian et al., 2013) . SRT3025 was tested in healthy male volunteers for the treatment of metabolic diseases but the trial was stopped by GlaxoSmithKline (GSK) due to unknown reasons (ClinicalTrials.gov Identifier: NCT01340911). Although NR administration increased blood NAD + levels and was well tolerated, it has not resulted in a striking improvement in any diseases so far (Dollerup et al., 2018; Dollerup et al., 2018; Katsyuba et al., 2020; Martens et al., 2018) . Oral administration of NMN was reported to be safe and feasible in healthy people. Furthermore, the trend toward decreased blood glucose levels in healthy participants after NMN treatment is a positive sign (Irie et al., 2020) . These findings suggest that challenges and barriers still exist in the translation of STACs to clinical practice. Questions to be answered include optimal dosage, duration of treatment, and long-term side effects. Therefore, future clinical studies need to be designed with good rationale to determine the dosage and administration duration for specific diseases (Gilmour et al., 2020; Lautrup et al., 2019) . Despite the abovementioned preclinical and clinical evidence for the effects of STACs, clinical trials, including osteoporosis as indication, are very limited. A randomized placebo-controlled trial showed that resveratrol treatment for 16 weeks significantly increased bone mass in elderly men with obesity (ClinicalTrials.gov Identifier: NCT01412645; Ornstrup et al., 2014) . A more recent randomized controlled trial also demonstrated that regular resveratrol supplementation improved BMD in postmenopausal women (Wong et al., 2020) . Further clinical trials to examine the effects of STACs alone or in combination with existing anti-osteoporosis drugs on the treatment of osteoporosis are of clinical interest. This extensive updated review reveals the important roles of sirtuins in diverse bone disorders. As sirtuins exert various biological effects on bone cells, they and their related underlying mechanisms are promising novel targets for the development of anti-osteoporotic therapies. Based on the current evidence, SIRT1 appears to be the most promising therapeutic target for the prevention of bone loss as it counteracts imbalanced bone remodeling through its actions on MSCs, osteoblasts, osteoclasts, and possibly hormone signaling pathways. In addition, the cumulative preclinical evidence of the anti-osteoporotic effects of SIRT1 activity enhancement has laid the foundation for many planned or ongoing clinical trials. Despite significant progress over the past few years, many research questions have yet to be answered. For example, it is important to know whether there is any discrepancy in terms of sirtuin activity with respect to sex and chronological age. Moreover, the biological functions of less studied sirtuins such as SIRT2, SIRT4, and SIRT5 in physiological and pathological conditions have not been adequately investigated. More in-depth mechanistic studies are expected to provide a better understanding of the effect of sirtuins on individual bone cell type by using tissue-specific deletion and transgenic animal models. From a clinical perspective, more specific and selective sirtuin activators with clearer pharmacokinetic and pharmacodynamic properties are needed to obtain more consistent and conclusive outcomes. The authors declare that they have no competing interests. QQL and YWL involved in concept and writing of the manuscript; QQL made the tables and created the figures; YWL, CYC, and QJ revised the manuscript; and QQL, CYC, QJ, and YWL performed analysis of the literature and critical discussion. Wayne Yuk-wai Lee https://orcid.org/0000-0002-0486-360X A complex secretory program orchestrated by the inflammasome controls paracrine senescence Skeletal involution by age-associated oxidative stress and its acceleration by loss of sex steroids Function of the SIRT 3 mitochondrial deacetylase in cellular physiology, cancer, and neurodegenerative disease Bone versus immune system The Sirtuin1 activator SRT3025 down-regulates sclerostin and rescues ovariectomy-induced bone loss and biomechanical deterioration in female mice Activation of Sirt1 decreases adipocyte formation during osteoblast differentiation of mesenchymal stem cells Association of oxidative stress with postmenopausal osteoporosis and the effects of hydrogen peroxide on osteoclast formation in human bone marrow cell cultures Clearance of p16 Ink4a-positive senescent cells delays ageing-associated disorders A phase II, randomized, placebo-controlled, double-blind, multi-dose study of SRT2104, a SIRT1 activator, in subjects with type 2 diabetes Mitochondria, oxidants, and aging. Cell SirT1 gain of function increases energy efficiency and prevents diabetes in mice FoxO proteins restrain osteoclastogenesis and bone resorption by attenuating H 2 O 2 accumulation Sir-two-homolog 2 (Sirt2) modulates peripheral myelination through polarity protein Par-3/ atypical protein kinase C (aPKC) signaling A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages The hallmarks of aging SirT1 regulates energy metabolism and response to caloric restriction in mice Slowing ageing by design: The rise of NAD+ and sirtuin-activating compounds Glucocorticoid-induced Osteoporosis Activation of SIRT3 by the NAD+ precursor nicotinamide riboside protects from noise-induced hearing loss Effects of tumor necrosis factor-alpha and interleukin-6 in elderly populations Prevalence of and risk factors for low bone mineral density and vertebral fractures in patients with systemic lupus erythematosus Mitochondrial free radical generation, oxidative stress, and aging Aging, cellular senescence, and cancer AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity Nonhomologous DNA end joining and alternative pathways to double-strand break repair Knockdown of SIRT7 enhances the osteogenic differentiation of human bone marrow mesenchymal stem cells partly via activation of the Wnt/β-catenin signaling pathway Activation of SIRT3 by resveratrol ameliorates cardiac fibrosis and improves cardiac function via the TGF-β/Smad3 pathway Developmental defects and p53 hyperacetylation in Sir2 homolog (SIRT1)-deficient mice Cyanidin chloride inhibits ovariectomy-induced osteoporosis by suppressing RANKLmediated osteoclastogenesis and associated signaling pathways Neuronal SIRT1 regulates endocrine and behavioral responses to calorie restriction Sirt1 is a regulator of bone mass and a repressor of Sost encoding for sclerostin, a bone formation inhibitor The senescence-associated secretory phenotype: The dark side of tumor suppression Sirtuin activators and inhibitors: Promises, achievements, and challenges A systematic review and economic evaluation of bisphosphonates for the prevention of fragility fractures Sirtuin 3 is required for osteogenic differentiation through maintenance of PGC-1ɑ-SOD2-mediated regulation of mitochondrial function A randomized placebo-controlled clinical trial of nicotinamide riboside in obese men: Safety, insulin-sensitivity, and lipid-mobilizing effects Aging and disease: Connections to sirtuins Regulation of cartilage-specific gene expression in human chondrocytes by SirT1 and nicotinamide phosphoribosyltransferase Silent information regulator (Sir) T1 inhibits NF-κB signaling to maintain normal skeletal remodeling Oxidative stress and vascular inflammation in aging SIRT1 is essential for oncogenic signaling by estrogen/estrogen receptor α in breast cancer Effect of estrogens on bone marrow adipogenesis and Sirt1 in aging C57BL/6J mice Reduced Sirtuin1 expression at the femoral neck in women who sustained an osteoporotic hip fracture Immunologic aspects of osteoporosis NAD+ augmentation restores mitophagy and limits accelerated aging in Werner syndrome NAD+ replenishment improves lifespan and healthspan in ataxia telangiectasia models via mitophagy and DNA repair Defective mitophagy in XPA via PARP-1 hyperactivation and NAD+/SIRT1 reduction Identification of senescent cells in the bone microenvironment Targeting cellular senescence prevents age-related bone loss in mice Protective effects of resveratrol on postmenopausal osteoporosis: Regulation of SIRT1-NF-κB signaling pathway Disorders of bone remodeling Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty Recent progress in the biology and physiology of sirtuins Potential involvement of SIRT1 in the pathogenesis of osteoarthritis through the modulation of chondrocyte gene expressions SIRT7 has a critical role in bone formation by regulating lysine acylation of SP7/ Osterix Glucose restriction inhibits skeletal myoblast differentiation by activating SIRT1 through AMPK-mediated regulation of Nampt SIRT3/SOD2 maintains osteoblast differentiation and bone formation by regulating mitochondrial stress Oxygen-derived free radicals stimulate osteoclastic bone resorption in rodent bone in vitro and in vivo A molecular mechanism for direct sirtuin activation by resveratrol Targeting NAD+ in translational research to relieve diseases and conditions of metabolic stress and ageing Mechanism of inhibition of bovine F1-ATPase by resveratrol and related polyphenols SIRT1 suppresses p53-dependent apoptosis by modulation of p21 in osteoblast-like MC3T3-E1 cells exposed to fluoride Calorie restriction-the SIR2 connection World-wide projections for hip fracture SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic β cells Mammalian sirtuins: Biological insights and disease relevance Control of osteoblast function and regulation of bone mass Hand cortical bone mass and its associations with radiographic joint damage and fractures in 50-70 year old female patients with rheumatoid arthritis: Cross sectional Oslo-Truro-Amsterdam (OSTRA) collaborative study Resveratrol prevents RANKL-induced osteoclast differentiation of murine osteoclast progenitor RAW 264.7 cells through inhibition of ROS production Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation Sirtuin-3 promotes adipogenesis, osteoclastogenesis, and bone loss in aging male mice Pharmacokinetics and tolerability of SRT 2104, a first-in-class small molecule activator of SIRT 1, after single and repeated oral administration in man Ionizing radiation induces cellular senescence of articular chondrocytes via negative regulation of SIRT1 by p38 kinase Ferulic acid, a natural polyphenol, protects against osteoporosis by activating SIRT1 and NF-κB in neonatal rats with glucocorticoid-induced osteoporosis Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan Sirt1 overexpression protects murine osteoblasts against TNF-αinduced injury in vitro by suppressing the NF-κB signaling pathway Small molecule SIRT1 activators for the treatment of aging and age-related diseases SIRT1-independent mechanisms of the putative sirtuin enzyme activators SRT1720 and SRT2183 Sirtuin 3 (SIRT3) maintains bone homeostasis by regulating AMPK-PGC-1β axis in mice NAD+ and sirtuins in aging and disease Effect of oral administration of nicotinamide mononucleotide on clinical parameters and nicotinamide metabolite levels in healthy Japanese men IL-6 in autoimmune disease and chronic inflammatory proliferative disease Coordination of PGC-1beta and iron uptake in mitochondrial biogenesis and osteoclast activation Sirtuin1 (Sirt1) promotes cortical bone formation by preventing β-catenin sequestration by FoxO transcription factors in osteoblast progenitors Reciprocal roles of DBC1 and SIRT1 in regulating estrogen receptor α activity and co-activator synergy Cartilage ablation of Sirt1 causes inhibition of growth plate chondrogenesis by hyperactivation of mTORC1 signaling SIRT2 deficiency prevents age-related bone loss in rats by inhibiting osteoclastogenesis The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms The sirtuin SIRT6 regulates lifespan in male mice SIRT6 protects against pathological damage caused by diet-induced obesity Regulation of SIRT6 protein levels by nutrient availability A novel role for the mitochondrial HTRA2/OMI protease in aging NAD+ homeostasis in health and disease Mutation in the silencing gene S/R4 can delay aging in S. cerevisiae The effect of resveratrol on normal and osteoarthritic chondrocyte metabolism DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age Sirtuin1 suppresses osteoclastogenesis by deacetylating FoxOs SOD2 and Sirt3 control osteoclastogenesis by regulating mitochondrial ROS SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity The mechanism of osteoclast differentiation induced by IL-1 Impact of the US Food and Drug Administration's safety-related announcements on the use of bisphosphonates after hip fracture Loss of Sirtuin 6 in osteoblast-lineage cells activates osteoclasts, resulting in osteopenia Forever young: SIRT3 a shield against mitochondrial meltdown, aging, and neurodegeneration Sirtuins and type 2 diabetes: Role in inflammation, oxidative stress, and mitochondrial function Mechanism of activation for the sirtuin 6 protein deacylase Mitochondrial superoxide in osteocytes perturbs canalicular networks in the setting of age-related osteoporosis The histone deacetylase SIRT1 controls male fertility in mice through regulation of hypothalamic-pituitary gonadotropin signaling Chromatin and beyond: The multitasking roles for SIRT6 SIRT6 suppresses pancreatic cancer through control of Lin28b Chemistry and biological activities of flavonoids: An overview NAD+ in brain aging and neurodegenerative disorders Cellular senescence and the biology of aging, disease, and frailty Gene expression profiling of glucocorticoid-inhibited osteoblasts The Sirt1 deacetylase modulates the insulin-like growth factor signaling pathway in mammals Glucocorticoid receptor and sequential P53 activation by dexamethasone mediates apoptosis and cell cycle arrest of osteoblastic MC3T3-E1 cells SIRT7 is a histone desuccinylase that functionally links to chromatin compaction and genome stability Estrogen receptor inhibits interleukin-6 gene expression by disruption of nuclear factor κB transactivation Mammalian Sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation Interactions between immune and bone cells: New insights with many remaining questions Denosumab or romosozumab therapy and risk of cardiovascular events in patients with primary osteoporosis: Systematic review and meta-analysis From estrogen-centric to aging and oxidative stress: A revised perspective of the pathogenesis of osteoporosis Bone marrow, cytokines, and bone remodeling-emerging insights into the pathophysiology of osteoporosis Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults Disruption of Sirt1 in chondrocytes causes accelerated progression of osteoarthritis under mechanical stress and during ageing in mice Mitochondria: More than just a powerhouse The mammalian SIR2α protein has a role in embryogenesis and gametogenesis SIRT6 stabilizes DNA-dependent protein kinase at chromatin for DNA double-strand break repair SIRT3 regulates progression and development of diseases of aging SRT 2104 extends survival of male mice on a standard diet and preserves bone and muscle mass Prevalence of osteoporosis and osteopenia among African Americans with early rheumatoid arthritis: The impact of ethnic-specific normative data Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet The prevalence of vertebral fractures in mild ankylosing spondylitis and their relationship to bone mineral density Intracellular and extracellular ATP coordinately regulate the inverse correlation between osteoclast survival and bone resorption Tumor necrosis factor-α polymorphism, bone strength phenotypes, and the risk of fracture in older women Sirtuin 6 in preosteoclasts suppresses age-and estrogen deficiency-related bone loss by stabilizing estrogen receptor α The RANKL/OPG system is activated in inflammatory bowel disease and relates to the state of bone loss Genomic instability and aging-like phenotype in the absence of mammalian SIRT6 The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling Metformin promotes the proliferation and differentiation of murine preosteoblast by regulating the expression of sirt6 and oct4 A senescent cell bystander effect: Senescence-induced senescence Structures of human sirtuin 3 complexes with ADPribose and with carba-NAD+ and SRT1720: Binding details and inhibition mechanism Trabecular bone mass and bone formation are preserved after limb immobilisation in p53 null mice Resveratrol increases bone mineral density and bone alkaline phosphatase in obese men: A randomized placebo-controlled trial Werner syndrome: Clinical features, pathogenesis and potential therapeutic interventions Ovarian steroid treatment blocks a postmenopausal increase in blood monocyte interleukin 1 release SIRT 7 clears the way for DNA repair Sirt6 cooperates with Blimp1 to positively regulate osteoclast differentiation Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span Changes in proinflammatory cytokine activity after menopause Sirt1 protects against high-fat diet-induced metabolic damage Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-γ Old age causes de novo intracortical bone remodeling and porosity in mice Honokiol, an activator of Sirtuin-3 (SIRT3) preserves mitochondria and protects the heart from doxorubicin-induced cardiomyopathy in mice Honokiol blocks and reverses cardiac hypertrophy in mice by activating mitochondrial Sirt3 A comprehensive overview on osteoporosis and its risk factors Natural polyphenols as sirtuin 6 modulators SIRT5 regulates the mitochondrial lysine succinylome and metabolic networks A suppressor of mating-type locus mutations in Saccharomyces cerevisiae: Evidence for and identification of cryptic mating-type loci Therapeutic approaches to bone diseases Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1 Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly A SIRT7-dependent acetylation switch of GABPβ1 controls mitochondrial function A human tRNA synthetase is a potent PARP1-activating effector target for resveratrol Disruption of the p53 gene results in preserved trabecular bone mass and bone formation after mechanical unloading Molecular mechanisms of aging-associated inflammation Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH The role of mammalian sirtuins in the regulation of metabolism, aging, and longevity The biochemistry of sirtuins Serum interleukin 6 is a major predictor of bone loss in women specific to the first decade past menopause Regulation of autophagy by ROS: Physiology and pathology TZDs and bone: A review of the recent clinical evidence Resveratrol mediated modulation of Sirt-1/Runx2 promotes osteogenic differentiation of mesenchymal stem cells: Potential role of Runx2 deacetylation SIRT1 functions as a negative regulator of eukaryotic poly (A) RNA transport SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease Cartilage-specific knockout of Sirt1 significantly reduces bone quality and catch-up growth efficiency Osteonecrosis of the jaw and the role of bisphosphonates: A critical review SIRT1 regulates differentiation of mesenchymal stem cells by deacetylating β-catenin Chemistry, pharmacology and health benefits of anthocyanins Osteocytic oxygen sensing controls bone mass through epigenetic regulation of sclerostin SIRT6 deficiency culminates in low-turnover osteopenia Mesenchymal progenitors in osteopenias of diverse pathologies: Differential characteristics in the common shift from osteoblastogenesis to adipogenesis SIRT6 regulates osteogenic differentiation of rat bone marrow mesenchymal stem cells partially via suppressing the nuclear factor-κB signaling pathway Overexpression of Sirt1 in mesenchymal stem cells protects against bone loss in mice by FOXO3a deacetylation and oxidative stress inhibition Higher-order unfolding of satellite heterochromatin is a consistent and early event in cell senescence SIRT1 regulation of apoptosis of human chondrocytes SIRT7-mediated ATM deacetylation is essential for its deactivation and DNA damage repair Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities Resveratrol and clinical trials: The crossroad from in vitro studies to human evidence The nutrient and energy sensor sirt1 regulates the hypothalamic-pituitary-adrenal (HPA) axis by altering the production of the prohormone convertase 2 (PC2) essential in the maturation of Corticotropin-releasing Hormone (CRH) from its prohormone in male rats Accelerated aging phenotype in mice with conditional deficiency for mitochondrial superoxide dismutase in the connective tissue Mitochondrial dysfunction as a cause of ageing Premature ageing in mice expressing defective mitochondrial DNA polymerase Resveratrol promotes osteogenesis of human mesenchymal stem cells by upregulating RUNX2 gene expression via the SIRT1/FOXO3A axis The effects of aging and sex steroid deficiency on the murine skeleton are independent and mechanistically distinct Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice Mitochondrial sirtuins and molecular mechanisms of aging Mitochondrial DNA deletion associated oxidative stress and severe male osteoporosis hSIR2SIRT1 functions as an NAD-dependent p53 deacetylase SIRT 7 promotes genome integrity and modulates non-homologous end joining DNA repair Cardiovascular effects of a novel SIRT1 activator, SRT2104, in otherwise healthy cigarette smokers Impairment of osteoblast differentiation due to proliferation-independent telomere dysfunction in mouse models of accelerated aging Sirt1 promotes osteogenic differentiation and increases alveolar bone mass via Bmi1 activation in mice The role of autophagy and mitophagy in bone metabolic disorders Protective effects of resveratrol on osteoporosis via activation of the SIRT1-NF-κB signaling pathway in rats Sirtuins, epigenetics and longevity IL-1 mediates TNF-induced osteoclastogenesis Regular supplementation with resveratrol improves bone mineral density in postmenopausal women: A randomised, placebo-controlled trial Exercise, inflammation and aging Inhibitory effect of Sirtuin6 (SIRT6) on osteogenic differentiation of bone marrow mesenchymal stem cells Targeting senescent cells enhances adipogenesis and metabolic function in old age Lethal cardiomyopathy in mice lacking transferrin receptor in the heart SIRT1/HERC4 locus associated with bisphosphonate-induced osteonecrosis of the jaw: An exome-wide association analysis Oxidative damage to osteoblasts can be alleviated by early autophagy through the endoplasmic reticulum stress pathway-Implications for the treatment of osteoporosis Upregulation of SIRT1 inhibits H 2 O 2 -induced osteoblast apoptosis via FoxO1/βcatenin pathway Modulation of NF-κB-dependent transcription and cell survival by the SIRT1 deacetylase SIRT1 directly regulates SOX2 to maintain self-renewal and multipotency in bone marrow-derived mesenchymal stem cells Structural basis of sirtuin 6 activation by synthetic small molecules Structural basis for the activation and inhibition of Sirtuin 6 by quercetin and its derivatives New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis Maldaptation of the link between inflammation and bone turnover may be a key determinant of osteoporosis Drugs targeting SIRT1, a new generation of therapeutics for osteoporosis and other bone related disorders? SIRT1 is a positive regulator of in vivo bone mass and a therapeutic target for osteoporosis SIRT1 is a positive regulator of the master osteoblast transcription factor, RUNX2 The prevalence of osteoporosis in China, a nationwide, multicenter DXA survey Phenotypic research on senile osteoporosis caused by SIRT6 deficiency Evidence for excessive osteoclast activation in SIRT6 null mice NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice The polyphenol resveratrol promotes skeletal growth in mice through a sirtuin 1-bone morphogenic protein 2 longevity axis Cellular senescence and the senescent secretory phenotype in age-related chronic diseases. Current Opinion in Clinical Nutrition & Metabolic Care Role of sirtuins in bone biology: Potential implications for novel therapeutic strategies for osteoporosis