Skip to main content

Roles of sirtuin family members in chronic obstructive pulmonary disease

Abstract

The globally increasing annual incidence of chronic obstructive pulmonary disease (COPD), a common chronic disease, poses a serious risk to public health. Although the exact mechanism underlying the pathogenesis of COPD remains unclear, a large number of studies have shown that its pathophysiology and disease course are closely related to oxidative stress, inflammation, apoptosis, autophagy, and aging. The key players involved in COPD include the sirtuin family of NAD-dependent deacetylases that comprise seven members (SIRT1–7) in mammals. Sirtuins play an important role in metabolic diseases, cell cycle control, proliferation, apoptosis, and senescence. Owing to differences in subcellular localization, sirtuins exhibit anisotropy. In this narrative review, we discuss the roles and molecular pathways of each member of the sirtuin family involved in COPD to provide novel insights into the prevention and treatment of COPD and how sirtuins may serve as adjuvants for COPD treatment.

Background

Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease that is not fully reversible. Currently, COPD is the fourth leading cause of death worldwide, and it is expected to rank even higher by 2030 [1]. COPD affects approximately 10% of individuals over the age of 45, and the disease is equally common among men and women in developed countries [2]. Smoking is a major risk factor for COPD [3]. Moreover, air pollution contributes to the increasing incidence of COPD [4]. The pathogenesis of COPD has not been fully understood. However, owing to its detrimental impact on public health, an increasing number of studies are being conducted on COPD.

Sirtuins are type III histone deacetylases (HDACs), which act on histone proteins present in the chromatin. The sirtuin family consists of seven members (SIRT1–7) in mammals. They exhibit different subcellular localizations in cells and play different roles based on the tissues and cells in which they are expressed. Sirtuins regulate cell metabolism, cell cycle, differentiation, autophagy/apoptosis, and senescence [5]. Owing to the correlation between the pathogenesis (described in the following section) and regulation of sirtuins, there have been many studies on the role of sirtuin family members in the regulation of COPD. Here, we review the roles and molecular pathways of each member of the sirtuin family involved in COPD to provide a reference for the prevention and treatment of this disease.

Pathogenesis of COPD

COPD is characterized by persistent obstruction in the airways, which is irreversible and progressive, leading to pulmonary ventilation dysfunction, and mainly manifests as chronic bronchitis (goblet cell proliferation and increased mucus secretion) [6] and emphysema (alveolar wall destruction, reduced gas diffusion) [7]. Most of these developments are progressive, leading to a gradual decline in lung function. The etiology of COPD is not yet fully understood, and its origin may be the culmination of long-term interactions between multiple environmental factors and the body’s endogenous factors [8]. Studies have shown that smoking is the greatest risk factor for COPD; however, many people who have never smoked also develop the disease, which may have its origin early in life [9]. Exposure of the fetus or child during infancy to adverse factors (e.g., cigarette exposure, maternal nutrition, birth weight, etc.) may lead to poor lung function [10]. Genetic factors, such as DNA methylation or changes in RNA expression patterns, can also influence the early development of COPD [11]. In addition, exposure to occupational dust, air pollution, infections, immune dysfunction, and aging are factors that lead to COPD [12].

The pathogenesis of COPD is a complex process involving oxidative stress, protease–antiprotease imbalance, and inflammation [13], which is related to the accelerated aging of lung cells [14]. Recent studies have shown that an important mechanism involves the downregulation of HDAC2, which plays an important role in inhibiting the expression of inflammatory genes [15]. Additionally, the endogenous oxidative stress caused by the activation of inflammatory cells in the lung owing to smoke inhalation is an important driving mechanism leading to the reduction in HDAC2 levels [16].

Airway remodeling, characterized by epithelial cell damage, small airway smooth muscle layer thickening, vascular proliferation, gland enlargement, increased mucus secretion, alveolar destruction, inflammatory cell infiltration, and epithelial–mesenchymal transition (EMT), is a characteristic pathological change of COPD [17]. Cigarette smoke stimulates the release and secretion of transforming growth factor (TGF)-β1, causes oxidative stress, and induces the differentiated bronchial epithelium to undergo EMT [18], which can induce airway remodeling and COPD [19].

COPD also accelerates aging of the lungs. Several studies have shown that the number of senescent cells in patients with COPD is increased [12, 20]. The incidence of COPD may be related to the aging of the human population, as the disease mainly affects the elderly, above the age of 65 years. The progression of airway obstruction in patients with COPD is gradual, and the decline in lung function accelerates with age, suggesting that COPD is involved in accelerating the aging process of the lungs [14, 21].

Therefore, it is important to identify the complex pathways involved in the pathogenesis of COPD. Sirtuins, as important anti-aging molecules, are involved in regulating oxidative stress and chronic inflammatory response, and may be involved in the occurrence and development of COPD; these activators may be potential therapeutic targets. From this perspective, sirtuins appear to be important target molecules and will be further discussed in this review.

Physiological function of sirtuins

The sirtuin protein family is an ancient family of proteins found across all eukaryotes. Sirtuin is an important gene-silencing complex that belongs to HDAC class III [22], and is considered an important regulator of longevity, anti-stress, anti-inflammation, and anti-aging [23]. However, its catalytic mechanism is different from that of other HDACs, as it requires nicotinamide adenine dinucleotide (NAD+) as a cofactor to catalyze the deacetylation of a specific substrate; this occurs by removing the acetyl-lysine of histones to disable ε-acetylamino, thereby inhibiting gene transcription [24, 25].

The sirtuin family was first discovered in yeast [26], and subsequently in higher eukaryotic nematodes [27] and Drosophila [28]. Sirtuins are involved in the regulation of multiple pathways, and play important roles in the regulation of DNA damage and pro-inflammatory gene transcription, which are closely related to longevity, apoptosis, and aging [29,30,31]. There are seven members in the mammalian sirtuin family, namely SIRT1–7 [32,33,34]. Studies have shown distinct subcellular localizations of each member of the sirtuin family. SIRT3, SIRT4, and SIRT5 are present in the cytoplasm and the mitochondria; SIRT6 is located in the nucleus; SIRT7 is localized in the nucleolus; and SIRT1 and SIRT2 can be found in both the nucleus and the cytoplasm [35, 36]. From the nucleus to the cytoplasm, sirtuins regulate a variety of cellular homeostasis mechanisms, playing different roles depending on the tissue and cell types in which they are expressed. Studies have shown that regulatory genes of the sirtuin family influence a variety of epigenetic factors, which can regulate the average life expectancy of the organism and affect their response to stress and toxicity [37]. Sirtuins also play important roles in other biological aspects such as metabolic diseases, and in the control of the cell cycle, cell proliferation, and aging, which could promote cell survival, apoptosis, or aging [37,38,39]. All seven members of the sirtuin family play different roles in COPD, which are discussed in further detail in the next section.

Sirtuins and COPD

SIRT1

SIRT1 was the first sirtuin discovered in mammals, which is the most widely studied sirtuin and is mainly found in the nucleus but also in the cytoplasm [40]. SIRT1 is an HDAC protein, which deacetylates many key regulatory proteins and transcription factors involved in DNA repair, inflammation, antioxidation, and cell aging, thereby regulating inflammatory responses, oxidative stress, autophagy/apoptosis, and aging [41]. Studies have shown that SIRT1 levels in the lung cells and peripheral blood mononuclear cells of patients with COPD are lower than those in healthy controls, which may be due to the increase in acetylation and the enhancement of inflammatory responses [42]. Therefore, SIRT1 may play an important role in inflammation regulation in the pathogenesis of COPD.

In human monocytes, SIRT1 is activated by Toll-like receptor 4, resulting in the deacetylation of p65/RelB and the termination of nuclear factor (NF)-ĸB-dependent gene transcription [43]. SIRT1 negatively regulates NF-κB, an inflammatory transcription factor, and inhibits gene transcription by deacetylating RELA/p65 [44]. Forkhead transcription factor 3a (FOXO3a) is a transcription factor of the forkhead family, which regulates the expression of multiple genes such as those involved in the cell cycle, differentiation, development, apoptosis, metabolism, autophagy, and longevity [45]. SIRT1 deacetylates FOXO3a, which plays an important role in cigarette smoke extract (CSE)-induced autophagy [46], and enhances the antioxidant response by inhibiting p53, inhibiting cell senescence and the NF-κB pathway, activating proliferator-activated receptor-coactivator-1 (PGC-1α), regulating mitochondrial dysfunction, and promoting cell survival under stress [47]. A recent study has shown that SIRT1 deficiency increases FOXO3 acetylation and exacerbates emphysema in CSE-induced mouse models, while activation of SIRT1 reduces FOXO3 acetylation and improves lung functions [48]. These important biological pathways seem to be involved in the development of COPD [49, 50]. Studies have shown that SIRT1 inhibits STAT3 activation in mouse embryonic cells, and acetylation is necessary for STAT3 activation [51]. As an important inflammatory mediator, STAT3 is involved in the regulation of the occurrence and development of COPD [52]. MicroRNAs are also involved in the regulation of SIRT1 expression and play a key role in oxidative stress and aging. MiR-34a reduces SIRT1 levels by activating the phosphoinositide-3 kinase (PI3K)–mammalian target of rapamycin (mTOR) pathway [53]. In turn, SIRT1 inhibits the mTOR signaling pathway and activates autophagy [54]. In COPD, miR-570 reduces SIRT1 levels by activating the p38 MAPK and activator protein (AP)-1 signaling pathways [55].

In addition, SIRT1 is involved in regulating EMT. Both in-vivo and in-vitro studies have shown that the upregulation of SIRT1 expression can inhibit activation of the TGF-β1/SMAD3 signaling pathway and weaken EMT, suggesting that SIRT1 plays an important role in the prevention of airway remodeling in COPD [56]. Transcriptome data obtained using microarray analysis of human lung fibroblasts, which are important participants in COPD fibrosis and airway remodeling, suggest that the transcription factor sterol regulatory element-binding protein 1 (SREBP1) is the main downstream regulator of SIRT1 in environmental COPD. Exposure of human lung fibroblasts to particulate matter (PM) leads to the activation of the SIRT1-SREBP1-PIR/NLRP3 inflammasome axis by inhibiting SIRT1, which can serve as therapeutic targets for PM-related COPD [57].

SIRT2

SIRT2 is the only sirtuin protein that exists in the cytoplasm (as its major localization), mitochondria, and nucleus. SIRT2 is involved in the regulation of many physiological processes, including the development of age-related diseases such as type 2 diabetes, cardiovascular diseases, neurodegenerative diseases, and COPD, which are all related to its genetic polymorphism [58,59,60,61,62]. SIRT2 may participate in the aging process via the deacetylation of α-tubulin [42]. In the cytoplasm, SIRT2 deacetylates p65, thereby regulating the expression of specific NF-κB-dependent genes [63].

Currently, the role of SIRT2 in the pathogenesis of COPD is unclear, with very few relevant studies. In a case–control study of COPD, 257 single nucleotide polymorphisms (SNPs) from 16 genes were selected and genotyped, and variations in SIRT2 were found to be associated with COPD susceptibility [64]. Another case–control study suggested that certain SIRT2 variants are important risk factors for COPD owing to the promotion of chronic systemic inflammation and oxidative stress [62]. These studies demonstrated that SIRT2 variants may play a role in the occurrence of COPD; however, further studies are required to clarify its biological function in COPD, which will help identify new therapeutic targets.

SIRT3

Evidence suggests that the impairment of mitochondrial structure and function owing to oxidative stress caused by long-term exposure to inhaled irritants, such as cigarette irritants, plays a key role in the development of COPD [65, 66]. Most mitochondrial proteins are acetylated and act as metabolic sensors, a reversible mechanism linking the metabolic state to mitochondrial function [67]. Acetyl-CoA mediates mitochondrial protein acetylation. SIRT3, SIRT4, and SIRT5 are mitochondrial proteins closely related to mitochondrial function and play an important role in mitochondria-related dysfunction during the progression of COPD.

SIRT3, the main deacetylase and the key regulator of mitochondrial activity, can deacetylate metabolic and respiratory enzymes to regulate mitochondrial function [68]. SIRT3 promotes mitochondrial redox balance by promoting NADPH production and activating manganese superoxide dismutase (MnSOD) to increase the production of reduced glutathione [69]. The lysine residue of MnSOD is deacetylated by SIRT3, which increases the enzymatic activity of MnSOD [70]. In addition, SIRT3 interacts with FOXO3a in the mitochondria, and the overexpression of SIRT3 increases the DNA-binding activity of FOXO3a, thereby stimulating the expression of the MnSOD gene [71]. Therefore, SIRT3 regulates mitochondrial oxidative stress via the upregulation and deacetylation of MnSOD. SIRT3 has also been reported to modulate NF-κB-mediated inflammation [72]. In COPD-related studies, SIRT3 activation may inhibit mitochondrial oxidative stress in airway epithelial cells by upregulation of MnSOD and prevent mitochondrial damage, which may provide a new strategy to slow down or prevent COPD [73].

Skeletal muscle dysfunction is one of the most serious extrapulmonary defects in patients with COPD, influenced by oxidative stress and energy metabolism disorder [74]. Mitochondrial damage plays an important role in the occurrence of skeletal muscle dysfunction during the progression of COPD [75]. SIRT3 is a downstream target of PGC-1α, which regulates the oxidation of mitochondrial fatty acids. The PGC-1α/SIRT3 signaling pathway is important in regulating mitochondrial metabolism and oxidative stress [76]. Studies have shown that curcumin can protect skeletal muscle mitochondria and alleviate skeletal muscle dysfunction by upregulating this signaling pathway in a COPD rat model [77].

SIRT4

SIRT4 is a key regulator of mitochondrial metabolic enzymes and antioxidant defense mechanisms. SIRT4 is present in the mitochondrial matrix of many organs, including the heart, kidney, brain, liver, and skeletal muscles [78,79,80]. However, the detailed mechanism underlying its enzymatic activity remains elusive. SIRT4 has ADP-ribosyltransferase [78], lipoamidase [81], deacylase [82], and substrate-specific deacetylase [83] activity. The enzyme activity and substrates of SIRT4 differ in different tissues and cells. For example, SIRT4 exhibits ADP-ribosyltransferase activity and inhibits insulin secretion in pancreatic beta cells [78]. It is also involved in the regulation of lipid oxidation ability in the liver and muscle mitochondrial function [84]. In addition, SIRT4 plays an important role in apoptosis, mitochondrial oxidative stress, and antioxidant response [85].

Thus far, there have been few studies on the role of SIRT4 in COPD. COPD is a complex inflammatory lung disease. Pulmonary endothelial dysfunction and vascular remodeling are closely related to its pathogenesis. Some studies have shown that SIRT4 overexpression can inhibit the expression of VCAM-1 and E-selectin in the inflammation of human pulmonary microvascular endothelial cells induced by CSE and can reduce the adhesion of monocytes to endothelial cells. In addition, SIRT4 overexpression can inhibit the degradation of IκBα, thereby inhibiting the nuclear translocation of NF-κB. However, it is still unclear whether SIRT4 is deacetylated via ADP-ribosylation or the NF-κB pathway. The activation of SIRT4 may contribute to the treatment of COPD-related endothelial inflammation and may serve as an effective target; however, elucidating its mechanism of action requires further research [86].

SIRT5

SIRT5 is located in the mitochondrial matrix and possesses deacetylase activity, but its activity is weak [87]. In addition, because of its ability to modify the negatively charged acyl group, the protein can be desuccinylated, deacylated, and deglutarated [87, 88]. SIRT5 plays a role in maintaining metabolism and cell homeostasis by regulating the activity of substrates in the metabolic process. SIRT5 can regulate myocardial energy metabolism and maintain the dynamic balance of the heart. SIRT5 participates in the formation and development of lung cancer as a tumor-specific promoter or inhibitor. SIRT5 can inhibit the occurrence and progression of neurodegenerative diseases [89].

Currently, there are limited studies on the role of SIRT5 in COPD. However, SIRT5 is involved in the regulation of metabolism, the stress response, and aging [90], and may, therefore, also be involved in the regulation of COPD. Studies have shown that SIRT5 can promote the deacetylation of FOXO3, thereby inhibiting the apoptosis of lung epithelial cells induced by CSE [91]. Thus, SIRT5 may prove to be a novel therapeutic target for smoking-related COPD.

SIRT6

SIRT6 is located in the nucleus and possesses deacetylase, ADP-ribosylase [92], and long-chain deacylase [93] activities. SIRT6 plays a key role in regulating DNA damage repair, telomere maintenance, genomic stability, inflammation, and metabolic homeostasis, and is associated with longevity [94].

A decrease in SIRT6 levels is observed in the lung and airway epithelial cells of patients with COPD [95, 96]. There are functional similarities between SIRT6 and SIRT1. SIRT6 is also involved in the regulation of autophagy. Interestingly, unlike SIRT1, SIRT6 inhibits cell senescence by inducing autophagy. SIRT6 overexpression can inhibit the senescence of human bronchial epithelial cells induced by CSE. Studies have shown that SIRT6 overexpression weakens autophagy through insulin-like growth factor (IGF)–Akt–mTOR signaling. SIRT6 is also regulated by miR34a, which activates the PI3K–mTOR pathway and leads to a decrease in SIRT6 expression [53] via a feedback loop. SIRT6 also inhibits the senescence induced by TGF-β via p21 proteasomal degradation and inhibits the secretion of IL-1β to prevent fibrosis [97]. SIRT6 is involved in the regulation of oxidative stress. Upon cigarette smoke stimulation, SIRT6 expression decreases, leading to NF-κB activation, reduction in telomere length, and reduction in the levels of β-catenin, VEGF, and NRF2, further increasing oxidative stress and thus accelerating aging and emphysema [12].

SIRT6 and SIRT1 play important roles in the manifestation of the extrapulmonary symptoms of COPD, such as peripheral skeletal muscle atrophy, a very common occurrence in COPD that seriously affects patients’ quality of life. In patients with COPD presenting with skeletal muscle atrophy, the expression level of phosphorylated H2AX, a molecule promoting DNA damage repair, is lower than that in healthy controls, which is correlated with the significantly lower protein levels of SIRT1 and SIRT6. These sirtuins may play a role in the regulation of diseases by repairing DNA damage and maintaining the stability of the genome [98].

SIRT7

SIRT7 is the least studied of the seven sirtuins to date. SIRT7 is located in the nucleolus; possesses deacetylase [99], desuccinylase [100], and long-chain deacylase [101] activities; and serves as a key mediator of several cellular activities. SIRT7 has been found to play a role in myocarditis [102], tumorigenesis [103], and apoptosis [104]. However, there is a dearth of studies elucidating the role of SIRT7 in COPD and the underlying molecular mechanisms. Nevertheless, recent research has shown that SIRT7 plays a role in the regulation of DNA damage repair [105], the cell cycle [106], and aging [107], suggesting that it also plays an important role in COPD pathogenesis. Therefore, further research on the role of SIRT7 in COPD is required to make a conclusive claim.

Potential application of sirtuin agonists in COPD

Thus far, numerous studies have been conducted on SIRT1 agonists with a potential role in the prevention and treatment of COPD. Here, we describe studies published in the recent 5 years on SIRT1 agonists and summarize them in Table 1.

Table 1 Molecular targets and functions of SIRT1 agonists

In a study of COPD rats, melatonin reduced airway inflammation by activating SIRT1 to inhibit the NLRP3 inflammasome and IL1β [108]. Melatonin also inhibited endoplasmic reticulum stress and improved alveolar epithelial cell apoptosis by upregulating SIRT1 expression [109]. Resveratrol is found in the peel of red fruit or red wine. It can protect the lungs by activating the SIRT1/PGC-1α pathway, regulating antioxidant enzymes, scavenging free radicals, reducing oxidative stress, and improving mitochondrial function [110]. Other studies have shown that hydrogen sulfide (H2S) can activate SIRT1, regulate EMT [56], weaken airway remodeling and mitochondrial dysfunction, and improve alveolar epithelial cell senescence and apoptosis [111]. Nucleosides isolated from Ophiocordyceps sinensis may inhibit COPD inflammation by activating SIRT1 to act on the NF-κB/P65 pathway in mice or mouse cells [112]. In rat models, curcumin can improve COPD by activating SIRT1 to regulate autophagy and endoplasmic reticulum stress [113]. Andrographolide may also inhibit extracellular signal-regulated kinase (ERK) signaling by activating SIRT1, preventing COPD mitochondrial dysfunction, inflammation, and oxidative stress [114]. Oxygen-regulated protein 150 (ORP150) is a member of the heat shock protein superfamily; melatonin and sodium hydrosulfide may attenuate apoptosis of human Bronchial epithelial (HBE) cells exposed to CSE by regulating endoplasmic reticulum stress response and autophagy through the SIRT1/ORP150 signaling pathway [115]. These agonists offer new possibilities for the treatment of COPD.

Compared with SIRT1, there is little research on the application of agonists in COPD with respect to other members of the sirtuin family. There have been some preclinical studies on SIRT3 agonists, including honokiol [116], melatonin [117], and berberine [118], which are being evaluated for use in the treatment of Alzheimer’s disease, cardiovascular disease, and non-alcoholic fatty liver disease. However, these agonists have not been tested for the treatment of COPD. Moreover, through study of the mechanism of sirtuins in COPD, it was revealed that sirtuins play important roles in COPD; hence, further study of sirtuin agonists is required such that they may be applied in clinical treatments in the future.

Conclusions

The sirtuin regulation mechanism of COPD is complex but mainly functions through inhibition of cellular senescence and inflammatory response, reduction of lung endothelial injury and oxidative stress, and regulation of autophagy to improve EMT and emphysema. Each sirtuin family member participates in the regulation of COPD through different pathways (as shown in Fig. 1).

Fig. 1
figure 1

Members of the sirtuin family are involved in the regulation of COPD. Briefly, SIRT1, SIRT2, SIRT6, and SIRT7 are localized in the nucleus. SIRT1, SIRT2 and SIRT6 inhibit the NF-κB signaling pathway to improve inflammatory response, SIRT1 and SIRT6 inhibit the TGF-β1 signaling pathway to alleviate EMT. SIRT1 can also regulate autophagy through the PI3K/mTOR signaling pathway. SIRT3, SIRT4, and SIRT5 are located in the cytoplasm. SIRT3 and SIRT1 improve oxidative stress response by interacting with PGC-1α, SIRT4 inhibits IκBα to improve pulmonary endothelial dysfunction, and SIRT5 and SIRT1 promote FOXO3a deacetylation to regulate apoptosis and autophagy

Among the members of the sirtuin family, SIRT1 and SIRT6 are the most widely studied. In particular, SIRT1 and its specific drug activators have been widely studied. However, other proteins in the family have been shown to play a role in the regulation of COPD via different pathways. COPD is highly detrimental to human health. Currently, there is no radical cure for advanced COPD, and the mortality rate is very high. Sirtuins may serve as adjuvants for COPD treatment.

In recent years, the sirtuin family has been extensively studied, leading to some remarkable discoveries. However, there have been very few studies on the impact of sirtuins on COPD. Therefore, it is important to further explore sirtuins as potential therapeutic targets of the disease. Specific drugs such as sirtuin-related agonists may prove to be an effective treatment strategy for COPD. However, owing to the abundant effects of sirtuins, such targeting may lead to potential side effects. Therefore, further studies are required to verify the roles and regulation pathways of sirtuin family members in COPD and to identify suitable targets for the treatment and prevention of COPD.

Availability of data and materials

Not applicable.

Abbreviations

COPD:

Chronic obstructive pulmonary disease

HDACs:

Histone deacetylases

EMT:

Epithelial–mesenchymal transition

NAD+:

Nicotinamide adenine dinucleotide

TLR:

Toll-like receptor

AP:

Activator protein

NF-κB:

Nuclear factor-κB

FOXO3a:

Forkhead transcription factor 3a

HPF:

Human lung fibroblast

PI3K:

Phosphoinositide-3 kinase

mTOR:

Mammalian target of rapamycin

SREBP1:

Sterol regulatory element-binding protein 1

PM:

Particulate matter

NLRP3:

NLR-family pyrin domain-containing protein 3

IL-1β:

Interleukin-1β

CHOP:

C/EBP homologous protein

ORP150:

Oxygen-regulated protein 150

HBE:

Human bronchial epithelial

H2S:

Hydrogen sulfide

TGF-β1:

Trans-forming growth factor-β1

PGC-1α:

Peroxisome proliferator-activated receptor γ coactivator 1α

MnSOD:

Manganese superoxide dismutase

HPMEC:

Human pulmonary microvascular endothelial cell

CSE:

Cigarette smoke extract

References

  1. Diaz-Guzman E, Mannino DM. Epidemiology and prevalence of chronic obstructive pulmonary disease. Clin Chest Med. 2014;35(1):7–16.

    PubMed  Google Scholar 

  2. Barnes PJ, Burney PG, Silverman EK, Celli BR, Vestbo J, Wedzicha JA, et al. Chronic obstructive pulmonary disease. Nat Rev Dis Primers. 2015;1:15076.

    PubMed  Google Scholar 

  3. Zhou JS, Li ZY, Xu XC, Zhao Y, Wang Y, Chen HP, et al. Cigarette smoke-initiated autoimmunity facilitates sensitisation to elastin-induced COPD-like pathologies in mice. Eur Respir J. 2020;56(3):2000404.

    CAS  PubMed  Google Scholar 

  4. Sood A, Assad NA, Barnes PJ, Churg A, Gordon SB, Harrod KS, et al. ERS/ATS workshop report on respiratory health effects of household air pollution. Eur Respir J. 2018;51(1):1700698.

    PubMed  PubMed Central  Google Scholar 

  5. Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M, Sinclair DA. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J Biol Chem. 2002;277(47):45099–107.

    CAS  PubMed  Google Scholar 

  6. Boucher RC. Muco-obstructive lung diseases. N Engl J Med. 2019;380(20):1941–53.

    CAS  PubMed  Google Scholar 

  7. Hogg JC, Timens W. The pathology of chronic obstructive pulmonary disease. Annu Rev Pathol. 2009;4:435–59.

    CAS  PubMed  Google Scholar 

  8. Huang X, Mu X, Deng L, Fu A, Pu E, Tang T, et al. The etiologic origins for chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2019;14:1139–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Salvi SS, Barnes PJ. Chronic obstructive pulmonary disease in non-smokers. Lancet (Lond, Engl). 2009;374(9691):733–43.

    Google Scholar 

  10. Narang I, Bush A. Early origins of chronic obstructive pulmonary disease. Semin Fetal Neonatal Med. 2012;17(2):112–8.

    PubMed  Google Scholar 

  11. Duijts L, Reiss IK, Brusselle G, de Jongste JC. Early origins of chronic obstructive lung diseases across the life course. Eur J Epidemiol. 2014;29(12):871–85.

    PubMed  Google Scholar 

  12. Barnes PJ. Senescence in COPD and its comorbidities. Annu Rev Physiol. 2017;79:517–39.

    CAS  PubMed  Google Scholar 

  13. Fischer BM, Pavlisko E, Voynow JA. Pathogenic triad in COPD: oxidative stress, protease-antiprotease imbalance, and inflammation. Int J Chron Obstruct Pulmon Dis. 2011;6:413–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ito K, Barnes PJ. COPD as a disease of accelerated lung aging. Chest. 2009;135(1):173–80.

    PubMed  Google Scholar 

  15. Barnes PJ. Role of HDAC2 in the pathophysiology of COPD. Annu Rev Physiol. 2009;71:451–64.

    CAS  PubMed  Google Scholar 

  16. Kirkham PA, Barnes PJ. Oxidative stress in COPD. Chest. 2013;144(1):266–73.

    CAS  PubMed  Google Scholar 

  17. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350(26):2645–53.

    CAS  PubMed  Google Scholar 

  18. Milara J, Peiró T, Serrano A, Cortijo J. Epithelial to mesenchymal transition is increased in patients with COPD and induced by cigarette smoke. Thorax. 2013;68(5):410–20.

    PubMed  Google Scholar 

  19. Hou W, Hu S, Li C, Ma H, Wang Q, Meng G, et al. Cigarette smoke induced lung barrier dysfunction, EMT, and tissue remodeling: a possible link between COPD and lung cancer. Biomed Res Int. 2019;2019:2025636.

    PubMed  PubMed Central  Google Scholar 

  20. Barnes PJ, Baker J, Donnelly LE. Cellular senescence as a mechanism and target in chronic lung diseases. Am J Respir Crit Care Med. 2019;200(5):556–64.

    CAS  PubMed  Google Scholar 

  21. Mercado N, Ito K, Barnes PJ. Accelerated ageing of the lung in COPD: new concepts. Thorax. 2015;70(5):482–9.

    PubMed  Google Scholar 

  22. Shore D, Squire M, Nasmyth KA. Characterization of two genes required for the position-effect control of yeast mating-type genes. EMBO J. 1984;3(12):2817–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Hwang JW, Yao H, Caito S, Sundar IK, Rahman I. Redox regulation of SIRT1 in inflammation and cellular senescence. Free Radic Biol Med. 2013;61:95–110.

    CAS  PubMed  Google Scholar 

  24. van de Ven RAH, Santos D, Haigis MC. Mitochondrial sirtuins and molecular mechanisms of aging. Trends Mol Med. 2017;23(4):320–31.

    PubMed  PubMed Central  Google Scholar 

  25. de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J. 2003;370(Pt 3):737–49.

    PubMed  PubMed Central  Google Scholar 

  26. Rine J, Herskowitz I. Four genes responsible for a position effect on expression from HML and HMR in Saccharomyces cerevisiae. Genetics. 1987;116(1):9–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Tissenbaum HA, Guarente L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature. 2001;410(6825):227–30.

    CAS  PubMed  Google Scholar 

  28. Rogina B, Helfand SL. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc Natl Acad Sci USA. 2004;101(45):15998–6003.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Krishnan V, Chow MZ, Wang Z, Zhang L, Liu B, Liu X, et al. Histone H4 lysine 16 hypoacetylation is associated with defective DNA repair and premature senescence in Zmpste24-deficient mice. Proc Natl Acad Sci USA. 2011;108(30):12325–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Ogiwara H, Ui A, Otsuka A, Satoh H, Yokomi I, Nakajima S, et al. Histone acetylation by CBP and p300 at double-strand break sites facilitates SWI/SNF chromatin remodeling and the recruitment of non-homologous end joining factors. Oncogene. 2011;30(18):2135–46.

    CAS  PubMed  Google Scholar 

  31. Mendes KL, Lelis DF, Santos SHS. Nuclear sirtuins and inflammatory signaling pathways. Cytokine Growth Factor Rev. 2017;38:98–105.

    CAS  PubMed  Google Scholar 

  32. Donmez G. The neurobiology of sirtuins and their role in neurodegeneration. Trends Pharmacol Sci. 2012;33(9):494–501.

    CAS  PubMed  Google Scholar 

  33. Kelly G. A review of the sirtuin system, its clinical implications, and the potential role of dietary activators like resveratrol: part 1. Altern Med Rev J Clin Ther. 2010;15(3):245–63.

    Google Scholar 

  34. Kelly GS. A review of the sirtuin system, its clinical implications, and the potential role of dietary activators like resveratrol: part 2. Altern Med Rev J Clin Ther. 2010;15(4):313–28.

    Google Scholar 

  35. Lin J, Sun B, Jiang C, Hong H, Zheng Y. Sirt2 suppresses inflammatory responses in collagen-induced arthritis. Biochem Biophys Res Commun. 2013;441(4):897–903.

    CAS  PubMed  Google Scholar 

  36. Haigis MC, Guarente LP. Mammalian sirtuins—emerging roles in physiology, aging, and calorie restriction. Genes Dev. 2006;20(21):2913–21.

    CAS  PubMed  Google Scholar 

  37. Michan S, Sinclair D. Sirtuins in mammals: insights into their biological function. Biochem J. 2007;404(1):1–13.

    CAS  PubMed  Google Scholar 

  38. Paraíso AF, Mendes KL, Santos SH. Brain activation of SIRT1: role in neuropathology. Mol Neurobiol. 2013;48(3):681–9.

    PubMed  Google Scholar 

  39. Rodriguez RM, Fraga MF. Aging and cancer: are sirtuins the link? Future Oncol (Lond, Engl). 2010;6(6):905–15.

    CAS  Google Scholar 

  40. Fang Y, Nicholl MB. Sirtuin 1 in malignant transformation: friend or foe? Cancer Lett. 2011;306(1):10–4.

    CAS  PubMed  Google Scholar 

  41. Rahman I, Kinnula VL, Gorbunova V, Yao H. SIRT1 as a therapeutic target in inflammaging of the pulmonary disease. Prev Med. 2012;54(Suppl):S20–8.

    CAS  PubMed  Google Scholar 

  42. Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I. SIRT1, an antiinflammatory and antiaging protein, is decreased in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177(8):861–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu TF, Yoza BK, El Gazzar M, Vachharajani VT, McCall CE. NAD+-dependent SIRT1 deacetylase participates in epigenetic reprogramming during endotoxin tolerance. J Biol Chem. 2011;286(11):9856–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Yang SR, Wright J, Bauter M, Seweryniak K, Kode A, Rahman I. Sirtuin regulates cigarette smoke-induced proinflammatory mediator release via RelA/p65 NF-kappaB in macrophages in vitro and in rat lungs in vivo: implications for chronic inflammation and aging. Am J Physiol Lung Cell Mol Physiol. 2007;292(2):L567–76.

    CAS  PubMed  Google Scholar 

  45. van der Horst A, Burgering BM. Stressing the role of FoxO proteins in lifespan and disease. Nat Rev Mol Cell Biol. 2007;8(6):440–50.

    PubMed  Google Scholar 

  46. Shi J, Yin N, Xuan LL, Yao CS, Meng AM, Hou Q. Vam3, a derivative of resveratrol, attenuates cigarette smoke-induced autophagy. Acta Pharmacol Sin. 2012;33(7):888–96.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Di Vincenzo S, Heijink IH, Noordhoek JA, Cipollina C, Siena L, Bruno A, et al. SIRT1/FoxO3 axis alteration leads to aberrant immune responses in bronchial epithelial cells. J Cell Mol Med. 2018;22(4):2272–82.

    PubMed  PubMed Central  Google Scholar 

  48. Jiang H, Jiang Y, Xu Y, Yuan D, Li Y. Bronchial epithelial SIRT1 deficiency exacerbates cigarette smoke induced emphysema in mice through the FOXO3/PINK1 pathway. Exp Lung Res. 2022. https://doi.org/10.1080/01902148.2022.2037169.

    Article  PubMed  Google Scholar 

  49. Edwards MR, Bartlett NW, Clarke D, Birrell M, Belvisi M, Johnston SL. Targeting the NF-kappaB pathway in asthma and chronic obstructive pulmonary disease. Pharmacol Ther. 2009;121(1):1–13.

    CAS  PubMed  Google Scholar 

  50. Ganesan S, Unger BL, Comstock AT, Angel KA, Mancuso P, Martinez FJ, et al. Aberrantly activated EGFR contributes to enhanced IL-8 expression in COPD airways epithelial cells via regulation of nuclear FoxO3A. Thorax. 2013;68(2):131–41.

    PubMed  Google Scholar 

  51. Nie Y, Erion DM, Yuan Z, Dietrich M, Shulman GI, Horvath TL, et al. STAT3 inhibition of gluconeogenesis is downregulated by SirT1. Nat Cell Biol. 2009;11(4):492–500.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kaur G, Batra S. Regulation of DNA methylation signatures on NF-κB and STAT3 pathway genes and TET activity in cigarette smoke extract-challenged cells/COPD exacerbation model in vitro. Cell Biol Toxicol. 2020;36(5):459–80.

    CAS  PubMed  Google Scholar 

  53. Baker JR, Vuppusetty C, Colley T, Papaioannou AI, Fenwick P, Donnelly L, et al. Oxidative stress dependent microRNA-34a activation via PI3Kα reduces the expression of sirtuin-1 and sirtuin-6 in epithelial cells. Sci Rep. 2016;6:35871.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, et al. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci USA. 2008;105(9):3374–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Baker JR, Vuppusetty C, Colley T, Hassibi S, Fenwick PS, Donnelly LE, et al. MicroRNA-570 is a novel regulator of cellular senescence and inflammaging. FASEB J. 2019;33(2):1605–16.

    CAS  PubMed  Google Scholar 

  56. Guan R, Wang J, Cai Z, Li Z, Wang L, Li Y, et al. Hydrogen sulfide attenuates cigarette smoke-induced airway remodeling by upregulating SIRT1 signaling pathway. Redox Biol. 2020;28:101356.

    CAS  PubMed  Google Scholar 

  57. Tien CP, Chen CH, Lin WY, Liu CS, Liu KJ, Hsiao M, et al. Ambient particulate matter attenuates Sirtuin1 and augments SREBP1-PIR axis to induce human pulmonary fibroblast inflammation: molecular mechanism of microenvironment associated with COPD. Aging. 2019;11(13):4654–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Liu T, Yang W, Pang S, Yu S, Yan B. Functional genetic variants within the SIRT2 gene promoter in type 2 diabetes mellitus. Diabetes Res Clin Pract. 2018;137:200–7.

    CAS  PubMed  Google Scholar 

  59. Hall JA, Dominy JE, Lee Y, Puigserver P. The sirtuin family’s role in aging and age-associated pathologies. J Clin Investig. 2013;123(3):973–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Porcelli S, Salfi R, Politis A, Atti AR, Albani D, Chierchia A, et al. Association between Sirtuin 2 gene rs10410544 polymorphism and depression in Alzheimer’s disease in two independent European samples. J Neural Transm (Vienna, Austria: 1996). 2013;120(12):1709–15.

    CAS  Google Scholar 

  61. Xia M, Yu JT, Miao D, Lu RC, Zheng XP, Tan L. SIRT2 polymorphism rs10410544 is associated with Alzheimer’s disease in a Han Chinese population. J Neurol Sci. 2014;336(1–2):48–51.

    CAS  PubMed  Google Scholar 

  62. Korytina GF, Akhmadishina LZ, Aznabaeva YG, Kochetova OV, Zagidullin NS, Kzhyshkowska JG, et al. Associations of the NRF2/KEAP1 pathway and antioxidant defense gene polymorphisms with chronic obstructive pulmonary disease. Gene. 2019;692:102–12.

    CAS  PubMed  Google Scholar 

  63. Rothgiesser KM, Erener S, Waibel S, Lüscher B, Hottiger MO. SIRT2 regulates NF-κB dependent gene expression through deacetylation of p65 Lys310. J Cell Sci. 2010;123(Pt 24):4251–8.

    CAS  PubMed  Google Scholar 

  64. Bakke PS, Zhu G, Gulsvik A, Kong X, Agusti AG, Calverley PM, et al. Candidate genes for COPD in two large data sets. Eur Respir J. 2011;37(2):255–63.

    CAS  PubMed  Google Scholar 

  65. Ahmad T, Sundar IK, Lerner CA, Gerloff J, Tormos AM, Yao H, et al. Impaired mitophagy leads to cigarette smoke stress-induced cellular senescence: implications for chronic obstructive pulmonary disease. FASEB J. 2015;29(7):2912–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Ito S, Araya J, Kurita Y, Kobayashi K, Takasaka N, Yoshida M, et al. PARK2-mediated mitophagy is involved in regulation of HBEC senescence in COPD pathogenesis. Autophagy. 2015;11(3):547–59.

    PubMed  PubMed Central  Google Scholar 

  67. Gao AW, Cantó C, Houtkooper RH. Mitochondrial response to nutrient availability and its role in metabolic disease. EMBO Mol Med. 2014;6(5):580–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, et al. Sirtuin 3, a new target of PGC-1alpha, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE. 2010;5(7):e11707.

    PubMed  PubMed Central  Google Scholar 

  69. Yu W, Dittenhafer-Reed KE, Denu JM. SIRT3 protein deacetylates isocitrate dehydrogenase 2 (IDH2) and regulates mitochondrial redox status. J Biol Chem. 2012;287(17):14078–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Shi H, Deng HX, Gius D, Schumacker PT, Surmeier DJ, Ma YC. Sirt3 protects dopaminergic neurons from mitochondrial oxidative stress. Hum Mol Genet. 2017;26(10):1915–26.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Jacobs KM, Pennington JD, Bisht KS, Aykin-Burns N, Kim HS, Mishra M, et al. SIRT3 interacts with the daf-16 homolog FOXO3a in the mitochondria, as well as increases FOXO3a dependent gene expression. Int J Biol Sci. 2008;4(5):291–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Chen CJ, Fu YC, Yu W, Wang W. SIRT3 protects cardiomyocytes from oxidative stress-mediated cell death by activating NF-κB. Biochem Biophys Res Commun. 2013;430(2):798–803.

    CAS  PubMed  Google Scholar 

  73. Zhang M, Zhang Y, Roth M, Zhang L, Shi R, Yang X, et al. Sirtuin 3 inhibits airway epithelial mitochondrial oxidative stress in cigarette smoke-induced COPD. Oxid Med Cell Longev. 2020;2020:7582980.

    PubMed  PubMed Central  Google Scholar 

  74. Pleguezuelos E, Esquinas C, Moreno E, Guirao L, Ortiz J, Garcia-Alsina J, et al. Muscular dysfunction in COPD: systemic effect or deconditioning? Lung. 2016;194(2):249–57.

    CAS  PubMed  Google Scholar 

  75. Meyer A, Zoll J, Charles AL, Charloux A, de Blay F, Diemunsch P, et al. Skeletal muscle mitochondrial dysfunction during chronic obstructive pulmonary disease: central actor and therapeutic target. Exp Physiol. 2013;98(6):1063–78.

    CAS  PubMed  Google Scholar 

  76. Rato L, Duarte AI, Tomás GD, Santos MS, Moreira PI, Socorro S, et al. Pre-diabetes alters testicular PGC1-α/SIRT3 axis modulating mitochondrial bioenergetics and oxidative stress. Biochem Biophys Acta. 2014;1837(3):335–44.

    CAS  PubMed  Google Scholar 

  77. Zhang M, Tang J, Li Y, Xie Y, Shan H, Chen M, et al. Curcumin attenuates skeletal muscle mitochondrial impairment in COPD rats: PGC-1α/SIRT3 pathway involved. Chem Biol Interact. 2017;277:168–75.

    CAS  PubMed  Google Scholar 

  78. Haigis MC, Mostoslavsky R, Haigis KM, Fahie K, Christodoulou DC, Murphy AJ, et al. SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic beta cells. Cell. 2006;126(5):941–54.

    CAS  PubMed  Google Scholar 

  79. Komlos D, Mann KD, Zhuo Y, Ricupero CL, Hart RP, Liu AY, et al. Glutamate dehydrogenase 1 and SIRT4 regulate glial development. Glia. 2013;61(3):394–408.

    PubMed  Google Scholar 

  80. Osborne B, Bentley NL, Montgomery MK, Turner N. The role of mitochondrial sirtuins in health and disease. Free Radic Biol Med. 2016;100:164–74.

    CAS  PubMed  Google Scholar 

  81. Mathias RA, Greco TM, Oberstein A, Budayeva HG, Chakrabarti R, Rowland EA, et al. Sirtuin 4 is a lipoamidase regulating pyruvate dehydrogenase complex activity. Cell. 2014;159(7):1615–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Anderson KA, Huynh FK, Fisher-Wellman K, Stuart JD, Peterson BS, Douros JD, et al. SIRT4 is a lysine deacylase that controls leucine metabolism and insulin secretion. Cell Metab. 2017;25(4):838-55.e15.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Michishita E, Park JY, Burneskis JM, Barrett JC, Horikawa I. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Mol Biol Cell. 2005;16(10):4623–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Nasrin N, Wu X, Fortier E, Feng Y, Bare OC, Chen S, et al. SIRT4 regulates fatty acid oxidation and mitochondrial gene expression in liver and muscle cells. J Biol Chem. 2010;285(42):31995–2002.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Han Y, Zhou S, Coetzee S, Chen A. SIRT4 and its roles in energy and redox metabolism in health, disease and during exercise. Front Physiol. 2019;10:1006.

    PubMed  PubMed Central  Google Scholar 

  86. Chen Y, Wang H, Luo G, Dai X. SIRT4 inhibits cigarette smoke extracts-induced mononuclear cell adhesion to human pulmonary microvascular endothelial cells via regulating NF-κB activity. Toxicol Lett. 2014;226(3):320–7.

    CAS  PubMed  Google Scholar 

  87. Du J, Zhou Y, Su X, Yu JJ, Khan S, Jiang H, et al. Sirt5 is a NAD-dependent protein lysine demalonylase and desuccinylase. Science (New York, NY). 2011;334(6057):806–9.

    CAS  Google Scholar 

  88. Tan M, Peng C, Anderson KA, Chhoy P, Xie Z, Dai L, et al. Lysine glutarylation is a protein posttranslational modification regulated by SIRT5. Cell Metab. 2014;19(4):605–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Kumar S, Lombard DB. Functions of the sirtuin deacylase SIRT5 in normal physiology and pathobiology. Crit Rev Biochem Mol Biol. 2018;53(3):311–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Sack MN, Finkel T. Mitochondrial metabolism, sirtuins, and aging. Cold Spring Harbor Perspect Biol. 2012;4(12):a013102.

    Google Scholar 

  91. Wang Y, Zhu Y, Xing S, Ma P, Lin D. SIRT5 prevents cigarette smoke extract-induced apoptosis in lung epithelial cells via deacetylation of FOXO3. Cell Stress Chaperones. 2015;20(5):805–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Van Meter M, Kashyap M, Rezazadeh S, Geneva AJ, Morello TD, Seluanov A, et al. SIRT6 represses LINE1 retrotransposons by ribosylating KAP1 but this repression fails with stress and age. Nat Commun. 2014;5:5011.

    PubMed  Google Scholar 

  93. Wang WW, Zeng Y, Wu B, Deiters A, Liu WR. A Chemical biology approach to reveal Sirt6-targeted Histone H3 sites in nucleosomes. ACS Chem Biol. 2016;11(7):1973–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Kugel S, Mostoslavsky R. Chromatin and beyond: the multitasking roles for SIRT6. Trends Biochem Sci. 2014;39(2):72–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Nakamaru Y, Vuppusetty C, Wada H, Milne JC, Ito M, Rossios C, et al. A protein deacetylase SIRT1 is a negative regulator of metalloproteinase-9. FASEB J. 2009;23(9):2810–9.

    CAS  PubMed  Google Scholar 

  96. Takasaka N, Araya J, Hara H, Ito S, Kobayashi K, Kurita Y, et al. Autophagy induction by SIRT6 through attenuation of insulin-like growth factor signaling is involved in the regulation of human bronchial epithelial cell senescence. J Immunol (Baltimore, Md: 1950). 2014;192(3):958–68.

    CAS  Google Scholar 

  97. Minagawa S, Araya J, Numata T, Nojiri S, Hara H, Yumino Y, et al. Accelerated epithelial cell senescence in IPF and the inhibitory role of SIRT6 in TGF-β-induced senescence of human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2011;300(3):L391-401.

    CAS  PubMed  Google Scholar 

  98. Lakhdar R, McGuinness D, Drost EM, Shiels PG, Bastos R, MacNee W, et al. Role of accelerated aging in limb muscle wasting of patients with COPD. Int J Chron Obstruct Pulmon Dis. 2018;13:1987–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Chen S, Blank MF, Iyer A, Huang B, Wang L, Grummt I, et al. SIRT7-dependent deacetylation of the U3–55k protein controls pre-rRNA processing. Nat Commun. 2016;7:10734.

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Li L, Shi L, Yang S, Yan R, Zhang D, Yang J, et al. SIRT7 is a histone desuccinylase that functionally links to chromatin compaction and genome stability. Nat Commun. 2016;7:12235.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Tong Z, Wang M, Wang Y, Kim DD, Grenier JK, Cao J, et al. SIRT7 is an RNA-activated protein lysine deacylase. ACS Chem Biol. 2017;12(1):300–10.

    CAS  PubMed  Google Scholar 

  102. Vakhrusheva O, Smolka C, Gajawada P, Kostin S, Boettger T, Kubin T, et al. Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ Res. 2008;102(6):703–10.

    CAS  PubMed  Google Scholar 

  103. Kiran S, Anwar T, Kiran M, Ramakrishna G. Sirtuin 7 in cell proliferation, stress and disease: Rise of the Seventh Sirtuin! Cell Signal. 2015;27(3):673–82.

    CAS  PubMed  Google Scholar 

  104. Blank MF, Grummt I. The seven faces of SIRT7. Transcription. 2017;8(2):67–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Tang M, Li Z, Zhang C, Lu X, Tu B, Cao Z, et al. SIRT7-mediated ATM deacetylation is essential for its deactivation and DNA damage repair. Sci Adv. 2019;5(3):eaav1118.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Lu YF, Xu XP, Lu XP, Zhu Q, Liu G, Bao YT, et al. SIRT7 activates p53 by enhancing PCAF-mediated MDM2 degradation to arrest the cell cycle. Oncogene. 2020;39(24):4650–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Kaiser A, Schmidt M, Huber O, Frietsch JJ, Scholl S, Heidel FH, et al. SIRT7: an influence factor in healthy aging and the development of age-dependent myeloid stem-cell disorders. Leukemia. 2020;34(8):2206–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Peng Z, Zhang W, Qiao J, He B. Melatonin attenuates airway inflammation via SIRT1 dependent inhibition of NLRP3 inflammasome and IL-1β in rats with COPD. Int Immunopharmacol. 2018;62:23–8.

    CAS  PubMed  Google Scholar 

  109. He B, Zhang W, Qiao J, Peng Z, Chai X. Melatonin protects against COPD by attenuating apoptosis and endoplasmic reticulum stress via upregulating SIRT1 expression in rats. Can J Physiol Pharmacol. 2019;97(5):386–91.

    CAS  PubMed  Google Scholar 

  110. Wang XL, Li T, Li JH, Miao SY, Xiao XZ. The effects of resveratrol on inflammation and oxidative stress in a rat model of chronic obstructive pulmonary disease. Molecules (Basel, Switzerland). 2017;22(9):1529.

    Google Scholar 

  111. Guan R, Cai Z, Wang J, Ding M, Li Z, Xu J, et al. Hydrogen sulfide attenuates mitochondrial dysfunction-induced cellular senescence and apoptosis in alveolar epithelial cells by upregulating sirtuin 1. Aging. 2019;11(24):11844–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Sun X, Dong Z, Li N, Feng X, Liu Y, Li A, et al. Nucleosides isolated from Ophiocordyceps sinensis inhibit cigarette smoke extract-induced inflammation via the SIRT1-nuclear factor-κB/p65 pathway in RAW2647 macrophages and in COPD mice. Int J Chronic Obstr Pulmon Dis. 2018;13:2821–32.

    CAS  Google Scholar 

  113. Tang F, Ling C. Curcumin ameliorates chronic obstructive pulmonary disease by modulating autophagy and endoplasmic reticulum stress through regulation of SIRT1 in a rat model. J Int Med Res. 2019;47(10):4764–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Zhang XF, Ding MJ, Cheng C, Zhang Y, Xiang SY, Lu J, et al. Andrographolide attenuates oxidative stress injury in cigarette smoke extract exposed macrophages through inhibiting SIRT1/ERK signaling. Int Immunopharmacol. 2020;81:106230.

    CAS  PubMed  Google Scholar 

  115. He B, Chen Q, Zhou D, Wang L, Liu Z. Role of reciprocal interaction between autophagy and endoplasmic reticulum stress in apoptosis of human bronchial epithelial cells induced by cigarette smoke extract. IUBMB Life. 2019;71(1):66–80.

    CAS  PubMed  Google Scholar 

  116. Ramesh S, Govindarajulu M, Lynd T, Briggs G, Adamek D, Jones E, et al. SIRT3 activator Honokiol attenuates β-amyloid by modulating amyloidogenic pathway. PLoS ONE. 2018;13(1):e0190350.

    PubMed  PubMed Central  Google Scholar 

  117. Zhai M, Li B, Duan W, Jing L, Zhang B, Zhang M, et al. Melatonin ameliorates myocardial ischemia reperfusion injury through SIRT3-dependent regulation of oxidative stress and apoptosis. J Pineal Res. 2017;63(2):e12419.

    Google Scholar 

  118. Xu X, Zhu XP, Bai JY, Xia P, Li Y, Lu Y, et al. Berberine alleviates nonalcoholic fatty liver induced by a high-fat diet in mice by activating SIRT3. FASEB J. 2019;33(6):7289–300.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was funded by the National Natural Science Foundation of China (Grant no. 81672297) and grants from the “Thirteenth Five-Year” Science and Technology Project of Jilin Provincial Department of Education (Grant no. JJKH20201093KJ).

Author information

Authors and Affiliations

Authors

Contributions

XYZ drafted the manuscript; JZ and XJL revised the manuscript and approved the final version; WL and JRZ made substantial contributions to the structure and literature review/data collection; CYL was responsible for all aspects of the work, ensuring the accuracy and completeness of the work. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jie Zhang or Xue-Jiao Lv.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, XY., Li, W., Zhang, JR. et al. Roles of sirtuin family members in chronic obstructive pulmonary disease. Respir Res 23, 66 (2022). https://doi.org/10.1186/s12931-022-01986-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-022-01986-y

Keywords