Fluoride Action Network

Abstract

This article has been Retracted.

Objective. To explore the function and mechanism of Sirt-1 in fluorine-induced liver injury.

Method. Fluorosis rats were first established. The fluorine content, pathological structure, collagen fibers, and fibrosis in liver tissues were tested through the fluoride ion selective electrode method, H&E, Masson, and Sirius red staining; alanine aminotransferase (ALT), aspartate aminotransferase (AST), interleukin 18 (IL-18), and tumor necrosis factor-a (TNF-a) levels in rat serum were also analyzed using ELISA kits. Then, the fluorosis cell model was built, which was also alleviated with NaF, Sirt-1 siRNAs, or endoplasmic reticulum stress (ERS) alleviator (4-PBA). CCK-8 also assessed cell proliferation; RT-qPCR or Western blots detect sirtuin-1 (Sirt-1), protein kinase R- (PKR-) like endoplasmic reticulum kinase (PERK), and endoplasmic reticulum stress (ERS) and apoptosis-related protein levels in liver tissue.

Results. Our results uncovered that fluorine exposure could aggravate the pathological damage and fibrosis of rat liver tissues and increase indicators related to liver injury. And fluoride exposure also could downregulate Sirt-1 and upregulate ERS-related proteins (PERK, 78-kD glucose-regulated protein (GRP-78), and activating transcription factor 6 (ATF6)) and apoptosis-related protein (caspase-3 and C/EBP-homologous protein (CHOP)) in rat liver tissues. Besides, we proved that fluoride exposure could suppress proliferation and enhances ERS and apoptotic pathways in AML12 cells by downregulating Sirt-1. Moreover, we revealed that ERS alleviator (4-PBA) could induce proliferation and prevent ERS and apoptosis in fluorine-exposed AML12 cells.

Conclusions. We suggested that fluorine exposure can induce hepatocyte ERS and apoptosis through downregulation of Sirt-1.


*Original article online at https://www.hindawi.com/journals/bmri/2022/7380324/

Excerpt:

1. Introduction

Fluorine exists in the environment in the form of fluoride [1]. And fluorine is a vital trace element present in humans and animals, mainly in bones and teeth [2, 3]. While long-term exposure to fluoride in the air, food, and water can lead to fluorosis, it can also cause dental fluorosis and fluorosis bone disease [4]. It has also been confirmed that chronic fluorosis can result in extensive pathological damage to the body [5]. Excessive intake of fluorine will cause morphological, functional, and metabolic changes in various organs, exposing soft tissues such as the liver, nerves, kidneys, blood vessels, and muscles to fluorine damage [6, 7]. The liver is the largest tissue organ in the body and can be involved in metabolism and blood production. Besides, the liver is the main organ for removing toxic substances from living organisms [8]. Long-term chronic fluoride exposure can lead to the accumulation of large amounts of fluoride in the liver, destroying its tissue morphology and affecting its normal physiological functions [9]. Several studies have indicated that sodium fluoride (NaF) can induce mitochondrial damage and promote hepatotoxicity and cellular damage [10, 11]. However, the mechanism of NaF-induced hepatotoxicity has not been clearly elucidated.

The endoplasmic reticulum (ER) is the site of protein synthesis, folding, and quality control [12]. During stressful conditions, unfolded and misfolded proteins can accumulate in the ER lumen, eventually causing ER stress (ERS) [13]. Research showed that ERS is associated with liver injury, and ERS-associated apoptosis is present throughout the process of liver injury [14]. Therefore, regulating ERS-associated apoptosis is very important to prevent liver injury.

Histone deacetylase (HDAC) can modify chromatin structure and regulate transcription factor activity [15]. And sirtuin-1 (Sirt-1) is a class III HDAC and can regulate biological processes including cellular metabolism, gene transcription, immune response, and glucose homeostasis through multiple deacetylation factors [16]. Sirt-1 has been reported to be associated with cell growth, apoptosis, senescence, autophagy, and other activities, which plays a key role in several diseases, such as neurodegenerative diseases, metabolic diseases, and cancer [1719]. Several researchers have also confirmed that Sirt-1 is relevant to liver injury [2022]. Besides, Sirt-1 also can exert a protective role against liver injury by suppressing ERS [23]. Recent study also revealed that Sirt-1 can weaken ERS by inhibiting the protein kinase R- (PKR-) like endoplasmic reticulum kinase- (PERK-) eIF2a-activating transcription factor 4 (ATF4) pathway, ultimately reducing ERS-induced apoptosis [24]. However, whether Sirt-1 can be involved in fluorine exposure-induced ERS in hepatocytes is not fully understood.

4. Discussion

Fluoride is widely present in the natural environment [33]. And long-term fluorine exposure can have certain toxic effects on the organism and cause significant hepatic pathological damage [34]. Currently, a study demonstrated that fluoride exposure could cause liver damage by the mitochondrial apoptosis pathway [9]. AST and ALT are the earliest and most sensitive indicators of the appearance of liver injury [35]. When liver tissue is necrotic or damaged, ALT and AST escape from hepatocytes and enter the bloodstream, significantly increasing serum ALT and AST activity [36]. Therefore, the increase of ALT and AST activity in serum reflects the degree of hepatocellular injury to a certain extent. In our study, we further proved that NaF treatment could induce necrosis and nuclear sequestration in hepatocytes and reduce intracellular organelles and swollen mitochondria in liver tissues of rats. Meanwhile, NaF also could elevate the levels of liver injury-related indicators (ALT and AST) and inflammatory indicators (IL-18, TNF-a) in rat serum. These results suggested that fluorine could induce liver injury in a dose-dependent manner.

Hepatic fibrosis (HF) is a repair response of the liver in response to chronic injury [37]. HF is also an intermediate stage in the progression of chronic liver disease to cirrhosis, which is a key stage in reversing the disease [38]. Late-stage HF may progress to irreversible cirrhosis [39]. And cirrhosis may further cause ascites, splenomegaly, formation of collateral circulation, upper gastrointestinal bleeding, and even death [40]. Our data further verified that fluorine exposure also could accelerate liver fibrosis in rats. Therefore, fluorine exposure can enhance liver injury and induce liver fibrosis.

Fluorosis is mainly associated with oxidative stress, hormonal regulation, and apoptosis [41]. Research showed that signaling pathways and related factors are relevant to fluorosis [42, 43]. To further explore the underlying mechanisms of fluorosis-induced liver injury, we investigated the effects of fluorine on hepatocyte ERS and apoptotic pathways. Apoptosis, a form of programmed cell death, can be induced by different toxic stimuli [44]. The literature reported that excess NaF could cause apoptosis in different cell types, including osteoblasts and human embryonic stem cells [45, 46]. And ERS is one of the key pathways of fluorine-induced apoptosis [47]. ERS acts as a cellular self-protection mechanism and normally has a role in protecting cells from damage. Adverse environments, such as oxidative stress and toxic stimuli, can accumulate unfolded and misfolded proteins in the ER, which can activate the unfolded protein response (UPR) [48]. UPR can maintain the balance of ER quantity and normal function in the body during ERS. Under stress, GRP78 can activate ERS through PERK, ATF6, and IRE1 [49]. While excessive ERS instead can activate ERS-associated apoptotic proteins such as CHOP, it can eventually trigger apoptosis [50]. In vivo study also showed that high fluorine concentrations can induce ERS and apoptosis in osteoblasts [51]. Our study further verified that fluoride exposure could upregulate ERS-and apoptosis-related proteins in liver tissues and AML12 cells. Thus, fluoride exposure could induce ERS and apoptosis in hepatic cells. Meanwhile, we discovered that ERS alleviator (4-PBA) could induce proliferation and inhibit ERS and apoptosis in fluorine-exposed AML12 cells, suggesting that fluorine exposure to hepatocyte ERS is critical.

More importantly, our data showed that fluoride exposure could prominently downregulate Sirt-1 in liver tissues and AML12 cells. Sirt-1 is a deacetylase that can regulate biological metabolism through deacetylation [52]. Besides, Sirt-1 has been reported to play key regulatory roles in physiological processes such as apoptosis, differentiation, oxidative stress, senescence, signaling, transcriptional regulation, and metabolic regulation through the regulation of histones, NF-kB, FOXO, and p53 [53, 54]. In recent years, studies confirmed that Sirt-1 is essential in liver-related diseases, such as liver transplantation [55], liver ischemia/reperfusion injury [56], liver fibrosis [57], fatty liver [58], alcoholic liver injury, and fibrosis [59]. At the same time, the role and mechanism of Sirt-1 in liver injury induced by fluoride exposure are unclear. Our results further indicated that Sirt-1 knockdown could further enhance the induction of ERS and apoptosis mediated by fluorine exposure in AML12 cells.

In our study, we first constructed fluorosis rat and cell models using NaF and clarified the influences of fluorine exposure on liver injury and fibrosis in rats. Besides, we explored the impacts of fluorine exposure on ERS- and apoptosis-related proteins in fluorosis rats and cells. Moreover, we further verified the action of Sirt-1 silencing and ERS alleviator (4-PBA) in fluorine-exposed rat liver tissues in vivo and AML12 cells in vitro. Therefore, the investigation of the protective mechanism of SIRT-1 against fluoride exposure-induced liver injury may provide a laboratory basis for the future clinical mitigation of fluorosis.

5. Conclusion

We demonstrated that fluorine exposure could induce hepatocyte injury through modulation of ERS and apoptotic pathways. Besides, Sirt-1 knockdown could further enhance the ERS and apoptotic processes in hepatocytes induced by fluoride exposure and enhance the toxic effects of NaF (Figure 6).

Data Availability

The datasets used and/or analyzed during the current study are available from the corresponding authors on reasonable request.

Conflicts of Interest

The authors declare no competing interests.

Authors’ Contributions

Yanlong Yu and Ling Li contributed equally to this work.

Acknowledgments

This work was supported by the Special Funds for the Central Government to Guide Local Science and Technology Development (grant no. QKZYD(2019)4008).

References

  1. J. Y. Yang, M. Wang, J. Lu et al., “Fluorine in the environment in an endemic fluorosis area in Southwest, China,” Environmental research, vol. 184, article 109300, 2020. View at: Publisher Site | Google Scholar
  2. S. Dehnen, L. L. Schafer, T. Lectka, and A. Togni, “Fluorine: a very special element and its very special impacts on chemistry,” The Journal of Organic Chemistry, vol. 86, no. 23, pp. 16213–16219, 2021. View at: Publisher Site | Google Scholar
  3. J. A. Horst, J. M. Tanzer, and P. M. Milgrom, “Fluorides and other preventive strategies for tooth decay,” Dental Clinics of North America, vol. 62, no. 2, pp. 207–234, 2018. View at: Publisher Site | Google Scholar
  4. J. Han, L. Kiss, H. Mei et al., “Chemical aspects of human and environmental overload with fluorine,” Chemical Reviews, vol. 121, no. 8, pp. 4678–4742, 2021. View at: Publisher Site | Google Scholar
  5. W. Wei, S. Pang, and D. Sun, “The pathogenesis of endemic fluorosis: research progress in the last 5 years,” Journal of Cellular and Molecular Medicine, vol. 23, no. 4, pp. 2333–2342, 2019. View at: Publisher Site | Google Scholar
  6. K. Dec, A. ?ukomska, D. Maciejewska et al., “The influence of fluorine on the disturbances of homeostasis in the central nervous system,” Biological Trace Element Research, vol. 177, no. 2, pp. 224–234, 2017. View at: Publisher Site | Google Scholar
  7. T. Nakamoto and H. R. Rawls, “Fluoride exposure in early life as the possible root cause of disease in later life,” The Journal of Clinical Pediatric Dentistry, vol. 42, no. 5, pp. 325–330, 2018. View at: Publisher Site | Google Scholar
  8. E. Trefts, M. Gannon, and D. H. Wasserman, “The liver,” Current Biology, vol. 27, no. 21, pp. R1147–r1151, 2017. View at: Publisher Site | Google Scholar
  9. H. Li, Z. Hao, L. Wang et al., “Dietary calcium alleviates fluorine-induced liver injury in rats by mitochondrial apoptosis pathway,” Biological Trace Element Research, vol. 200, no. 1, pp. 271–280, 2022. View at: Publisher Site | Google Scholar
  10. R. Mitta, S. Duddu, R. Y. Pulala, P. Bhupalam, V. Mandlem, and A. Konde, “Mitigative effect of <i>Momordica cymbalaria</i> fruit extract against sodium fluoride induced hepatotoxicity in Wistar male albino rats,” Journal of Basic and Clinical Physiology and Pharmacology, vol. 32, no. 2, pp. 79–87, 2020. View at: Publisher Site | Google Scholar
  11. C. Song, J. Zhao, B. Fu et al., “Melatonin-mediated upregulation of Sirt3 attenuates sodium fluoride-induced hepatotoxicity by activating the MT1-PI3K/AKT-PGC-1? signaling pathway,” Free Radical Biology & Medicine, vol. 112, pp. 616–630, 2017. View at: Publisher Site | Google Scholar
  12. Q. M. Zhai, B. Li, X. N. He et al., “Endoplasmic reticulum and its significance in periodontal disease,” The Chinese Journal of Dental Research, vol. 24, no. 2, pp. 79–84, 2021. View at: Publisher Site | Google Scholar
  13. Z. Qi and L. Chen, “Endoplasmic reticulum stress and autophagy,” Advances in Experimental Medicine and Biology, vol. 1206, pp. 167–177, 2019. View at: Publisher Site | Google Scholar
  14. W. J. Qian and Q. H. Cheng, “Endoplasmic reticulum stress-mediated apoptosis signal pathway is involved in sepsis-induced liver injury,” International Journal of Clinical and Experimental Pathology, vol. 10, no. 9, pp. 9990–9997, 2017. View at: Google Scholar
  15. L. M. Zhao and J. H. Zhang, “Histone deacetylase inhibitors in tumor immunotherapy,” Current Medicinal Chemistry, vol. 26, no. 17, pp. 2990–3008, 2019. View at: Publisher Site | Google Scholar
  16. C. F. Aylwin and A. Lomniczi, “Sirtuin (SIRT)-1: at the crossroads of puberty and metabolism,” Current opinion in endocrine and metabolic research, vol. 14, pp. 65–72, 2020. View at: Publisher Site | Google Scholar
  17. L. Liu, G. Xia, P. Li, Y. Wang, and Q. Zhao, “Sirt-1 regulates physiological process and exerts protective effects against oxidative stress,” BioMed Research International, vol. 2021, Article ID 5542545, 12 pages, 2021. View at: Publisher Site | Google Scholar
  18. D. R. Machin, Y. Auduong, V. R. Gogulamudi et al., “Lifelong SIRT-1 overexpression attenuates large artery stiffening with advancing age,” Aging, vol. 12, no. 12, pp. 11314–11324, 2020. View at: Publisher Site | Google Scholar
  19. A. Garten, T. Grohmann, K. Kluckova, G. G. Lavery, W. Kiess, and M. Penke, “Sorafenib-induced apoptosis in hepatocellular carcinoma is reversed by SIRT1,” International Journal of Molecular Sciences, vol. 20, no. 16, p. 4048, 2019. View at: Publisher Site | Google Scholar
  20. T. Dusabimana, S. R. Kim, H. J. Kim, S. W. Park, and H. Kim, “Nobiletin ameliorates hepatic ischemia and reperfusion injury through the activation of SIRT-1/FOXO3a-mediated autophagy and mitochondrial biogenesis,” Experimental & Molecular Medicine, vol. 51, no. 4, pp. 1–16, 2019. View at: Publisher Site | Google Scholar
  21. C. Zeng, X. Hu, W. He et al., “Hypothermic machine perfusion ameliorates inflammation during ischemia-reperfusion injury via sirtuin-1-mediated deacetylation of nuclear factor-?B p65 in rat livers donated after circulatory death,” Molecular Medicine Reports, vol. 16, no. 6, pp. 8649–8656, 2017. View at: Publisher Site | Google Scholar
  22. H. R. Jing, F. W. Luo, X. M. Liu, X. F. Tian, and Y. Zhou, “Fish oil alleviates liver injury induced by intestinal ischemia/reperfusionviaAMPK/SIRT-1/autophagy pathway,” World Journal of Gastroenterology, vol. 24, no. 7, pp. 833–843, 2018. View at: Publisher Site | Google Scholar
  23. J. Lee, S. W. Hong, S. E. Park et al., “Exendin-4 attenuates endoplasmic reticulum stress through a SIRT1-dependent mechanism,” Cell Stress & Chaperones, vol. 19, no. 5, pp. 649–656, 2014. View at: Publisher Site | Google Scholar
  24. Z. Lin, C. Teng, L. Ni et al., “Echinacoside upregulates Sirt1 to suppress endoplasmic reticulum stress and inhibit extracellular matrix degradation in vitro and ameliorates osteoarthritis in vivo,” Oxidative Medicine and Cellular Longevity, vol. 2021, Article ID 3137066, 21 pages, 2021. View at: Publisher Site | Google Scholar
  25. B. E. Phillips, L. Lantier, C. Engman et al., “Improvement in insulin sensitivity and prevention of high fat diet-induced liver pathology using a CXCR2 antagonist,” Cardiovascular Diabetology, vol. 21, no. 1, p. 130, 2022. View at: Publisher Site | Google Scholar
  26. H. Huo, H. Wu, F. Ma et al., “N-acetyl-L-cysteine ameliorates hepatocyte pyroptosis of dog type 1 diabetes mellitus via suppression of NLRP3/NF-?B pathway,” Life Sciences, vol. 306, p. 120802, 2022. View at: Publisher Site | Google Scholar
  27. K. Zhang, L. Lin, Y. Zhu, N. Zhang, M. Zhou, and Y. Li, “Saikosaponin d alleviates liver fibrosis by negatively regulating the ROS/NLRP3 inflammasome through activating the ER? pathway,” Frontiers in Pharmacology, vol. 13, article 894981, 2022, Published 2022 May 25. View at: Publisher Site | Google Scholar
  28. W. Yu and Q. Mao, “Inhibition of TRAF1 protects renal tubular epithelial cells against hypoxia/reoxygenation injury,” Journal of Mens Health, vol. 17, no. 3, pp. 167–173, 2021. View at: Publisher Site | Google Scholar
  29. L. Carreres, M. Mercey-Ressejac, K. Kurma et al., “Chronic intermittent hypoxia increases cell proliferation in hepatocellular carcinoma,” Cells, vol. 11, no. 13, p. 2051, 2022. View at: Publisher Site | Google Scholar
  30. X. F. Chen, Y. Wang, S. Ji et al., “Hepatoprotective efficacy and interventional mechanism of Qijia Rougan decoction in liver fibrosis,” Frontiers in Pharmacology, vol. 13, article 911250, 2022. View at: Publisher Site | Google Scholar
  31. Z. Peng, M. Li, Y. Wang et al., “Self-assembling Imageable silk hydrogels for the focal treatment of osteosarcoma,” Developmental Biology, vol. 10, article 698282, 2022. View at: Publisher Site | Google Scholar
  32. G. D. Smedley, K. E. Walker, and S. H. Yuan, “The role of PERK in understanding development of neurodegenerative diseases,” International Journal of Molecular Sciences, vol. 22, no. 15, p. 8146, 2021. View at: Publisher Site | Google Scholar
  33. N. R. Johnston and S. A. Strobel, “Principles of fluoride toxicity and the cellular response: a review,” Archives of Toxicology, vol. 94, no. 4, pp. 1051–1069, 2020. View at: Publisher Site | Google Scholar
  34. A. G. Zhukova, N. N. Mikhailova, T. K. Yadykina et al., “Experimental studies of intracellular liver protective mechanisms in development of chronic fluorine intoxication,” Meditsina Truda i Promyshlennaia Ekologiia, vol. 5, pp. 21–24, 2016. View at: Google Scholar
  35. L. Xu, Y. Yu, R. Sang, J. Li, B. Ge, and X. Zhang, “Protective effects of taraxasterol against ethanol-induced liver injury by regulating CYP2E1/Nrf2/HO-1 and NF-?B signaling pathways in mice,” Oxidative Medicine and Cellular Longevity, vol. 2018, Article ID 8284107, 11 pages, 2018. View at: Publisher Site | Google Scholar
  36. R. Yuan, X. Tao, S. Liang et al., “Protective effect of acidic polysaccharide from Schisandra chinensis on acute ethanol-induced liver injury through reducing CYP2E1-dependent oxidative stress,” Biomedicine & Pharmacotherapy, vol. 99, pp. 537–542, 2018. View at: Publisher Site | Google Scholar
  37. L. Caballería, P. Torán, and J. Caballería, “Markers of hepatic fibrosis,” Medicina Clínica, vol. 150, no. 8, pp. 310–316, 2018. View at: Publisher Site | Google Scholar
  38. R. M. Dawood, M. A. el-Meguid, G. M. Salum, and M. K. el Awady, “Key players of hepatic fibrosis,” Journal of Interferon & Cytokine Research, vol. 40, no. 10, pp. 472–489, 2020. View at: Publisher Site | Google Scholar
  39. S. Khan and R. Saxena, “Regression of hepatic fibrosis and evolution of cirrhosis: a concise review,” Advances in Anatomic Pathology, vol. 28, no. 6, pp. 408–414, 2021. View at: Publisher Site | Google Scholar
  40. P. Ginès, A. Krag, J. G. Abraldes, E. Solà, N. Fabrellas, and P. S. Kamath, “Liver cirrhosis,” The Lancet, vol. 398, no. 10308, pp. 1359–1376, 2021. View at: Publisher Site | Google Scholar
  41. W. Li, S. Dong, Q. Chen, C. Chen, and Z. Dong, “Selenium may suppress peripheral blood mononuclear cell apoptosis by modulating HSP70 and regulate levels of SIRT1 through reproductive hormone secretion and oxidant stress in women suffering fluorosis,” European Journal of Pharmacology, vol. 878, article 173098, 2020. View at: Publisher Site | Google Scholar
  42. L. Qiao, X. Liu, Y. He et al., “Progress of signaling pathways, stress pathways and epigenetics in the pathogenesis of skeletal fluorosis,” International Journal of Molecular Sciences, vol. 22, no. 21, p. 11932, 2021. View at: Publisher Site | Google Scholar
  43. L. Ma, R. Zhang, D. Li, T. Qiao, and X. Guo, “Fluoride regulates chondrocyte proliferation and autophagy via PI3K/AKT/mTOR signaling pathway,” Chemico-Biological Interactions, vol. 349, article 109659, 2021. View at: Publisher Site | Google Scholar
  44. E. Obeng, “Apoptosis (programmed cell death) and its signals – a review,” Brazilian Journal of Biology, vol. 81, no. 4, pp. 1133–1143, 2021. View at: Publisher Site | Google Scholar
  45. X. Li, L. Meng, F. Wang, X. Hu, and Y. Yu, “Sodium fluoride induces apoptosis and autophagy via the endoplasmic reticulum stress pathway in MC3T3-E1 osteoblastic cells,” Molecular and Cellular Biochemistry, vol. 454, no. 1-2, pp. 77–85, 2019. View at: Publisher Site | Google Scholar
  46. T. D. Nguyen Ngoc, Y. O. Son, S. S. Lim et al., “Sodium fluoride induces apoptosis in mouse embryonic stem cells through ROS- dependent and caspase- and JNK-mediated pathways,” Toxicology and Applied Pharmacology, vol. 259, no. 3, pp. 329–337, 2012. View at: Publisher Site | Google Scholar
  47. C. Hetz, K. Zhang, and R. J. Kaufman, “Mechanisms, regulation and functions of the unfolded protein response,” Nature Reviews. Molecular Cell Biology, vol. 21, no. 8, pp. 421–438, 2020. View at: Publisher Site | Google Scholar
  48. J. Ren, Y. Bi, J. R. Sowers, C. Hetz, and Y. Zhang, “Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases,” Nature Reviews. Cardiology, vol. 18, no. 7, pp. 499–521, 2021. View at: Publisher Site | Google Scholar
  49. W. Li, T. Cao, C. Luo et al., “Crosstalk between ER stress, NLRP3 inflammasome, and inflammation,” Applied Microbiology and Biotechnology, vol. 104, no. 14, pp. 6129–6140, 2020. View at: Publisher Site | Google Scholar
  50. A. V. Cybulsky, “Endoplasmic reticulum stress, the unfolded protein response and autophagy in kidney diseases,” Nature Reviews. Nephrology, vol. 13, no. 11, pp. 681–696, 2017. View at: Publisher Site | Google Scholar
  51. L. Liu, Y. Zhang, H. Gu, K. Zhang, and L. Ma, “Fluorosis induces endoplasmic reticulum stress and apoptosis in osteoblasts In Vivo,” Biological Trace Element Research, vol. 164, no. 1, pp. 64–71, 2015. View at: Publisher Site | Google Scholar
  52. K. Devi, N. Singh, and A. S. Jaggi, “Dual role of sirtuin 1 in inflammatory bowel disease,” Immunopharmacology and Immunotoxicology, vol. 42, no. 5, pp. 385–391, 2020. View at: Publisher Site | Google Scholar
  53. S. H. Lee, J. H. Lee, H. Y. Lee, and K. J. Min, “Sirtuin signaling in cellular senescence and aging,” BMB Reports, vol. 52, no. 1, pp. 24–34, 2019. View at: Publisher Site | Google Scholar
  54. W. Wang, W. Sun, Y. Cheng, Z. Xu, and L. Cai, “Role of sirtuin-1 in diabetic nephropathy,” Journal of Molecular Medicine, vol. 97, no. 3, pp. 291–309, 2019. View at: Publisher Site | Google Scholar
  55. U. Scheuermann, E. R. Seyferth, N. Abraham et al., “Sirtuin-1 expression and activity is diminished in aged liver grafts,” Scientific Reports, vol. 10, no. 1, p. 11860, 2020. View at: Publisher Site | Google Scholar
  56. H. Yan, Y. Jihong, Z. Feng et al., “Sirtuin 1-mediated inhibition of p66shc expression alleviates liver ischemia/reperfusion injury,” Critical Care Medicine, vol. 42, no. 5, pp. e373–e381, 2014. View at: Publisher Site | Google Scholar
  57. X. Luo, Y. Bai, S. He et al., “Sirtuin 1 ameliorates defenestration in hepatic sinusoidal endothelial cells during liver fibrosis via inhibiting stress-induced premature senescence,” Cell Proliferation, vol. 54, no. 3, article e12991, 2021. View at: Publisher Site | Google Scholar
  58. R. B. Ding, J. Bao, and C. X. Deng, “Emerging roles of SIRT1 in fatty liver diseases,” International Journal of Biological Sciences, vol. 13, no. 7, pp. 852–867, 2017. View at: Publisher Site | Google Scholar
  59. T. Ramirez, Y. M. Li, S. Yin et al., “Aging aggravates alcoholic liver injury and fibrosis in mice by downregulating sirtuin 1 expression,” Journal of Hepatology, vol. 66, no. 3, pp. 601–609, 2017. View at: Publisher Site | Google Scholar

Copyright © 2022 Yanlong Yu et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.