Colchicine Ameliorates Dilated Cardiomyopathy Via SIRT2-Mediated Suppression of NLRP3 Inflammasome Activation

Xuan Sun, Junfeng Duan, Chenyi Gong, Yuting Feng, Jiaxin Hu, Rong Gu, Biao Xu, Xuan Sun, Junfeng Duan, Chenyi Gong, Yuting Feng, Jiaxin Hu, Rong Gu, Biao Xu

Abstract

Background Dilated cardiomyopathy remains a leading cause of heart failure worldwide. Immune inflammation response is recognized as a significant player in the progression of heart failure; however, immunomodulatory strategies remain a long-term challenge. Colchicine, a potent anti-inflammatory drug, has many benefits in ischemic cardiovascular events, but its role in nonischemic heart failure remains unclear. Methods and Results Doxorubicin administration was used to establish a murine dilated cardiomyopathy model, and colchicine or saline was orally given. At the end point, cardiac function and fibrosis were measured to investigate the effects of colchicine. Inflammatory cytokine levels, neutrophil recruitment, and NLRP3 (NOD-like receptor protein 3) inflammasome activation were detected to evaluate the inflammatory response. Furthermore, to examine the downstream target of colchicine, SIRT2 (Sirtuin 2) was pharmacologically inhibited in vitro; thus, changes in the NLRP3 inflammasome were detected by immunoblotting. These results showed that murine cardiac function was significantly improved and fibrosis was significantly alleviated after colchicine treatment. Moreover, the infiltration of neutrophils and the levels of inflammatory cytokines in the failing myocardium were both decreased by colchicine treatment. Mechanistically, colchicine upregulated the expression of SIRT2, leading to the inactivation of the NLRP3 inflammasome in an NLRP3 deacetylated manner. Conversely, the inhibition of SIRT2 attenuated the suppressive effect of colchicine on NLRP3 inflammasome activation. Conclusions This study indicated that colchicine could be a promising therapeutic candidate for dilated cardiomyopathy and other nonischemic heart failure associated with the inflammatory response.

Keywords: NLRP3 inflammasome; SIRT2; colchicine; dilated cardiomyopathy; inflammation.

Figures

Figure 1. Colchicine treatment significantly improved murine…
Figure 1. Colchicine treatment significantly improved murine cardiac functions.
A through C, Cardiac function is analyzed by 2‐dimensional murine echocardiography, left ventricular (LV) ejection fraction, LV fractional shortening, LV internal dimensions at end‐diastole, and LV internal dimensions at end‐systole values are measured at the end point of experiment (n=10 each group). D, Kaplan‐Meier survival curve for each group mice followed for 4 weeks with or without doxorubicin challenge (n=25 each group). E, The changes of body weight from beginning to end (n=6 each group). F, Relative expression of ANP (atrial natriuretic peptide) mRNA and BNP (brain natriuretic peptide) mRNA are normalized to GAPDH mRNA in murine cardiac tissues at the end point of experiment. G and H, The representative hematoxylin and eosin staining (G) and Sirius red staining (H) of hearts in each group are shown. Quantification of interstitial fibrosis (% area) in hearts is shown on the right; n=8 each group. Five images per mouse are evaluated. Scale bar=50 μm. Data are shown as mean±SEM. Data of (B, C, and E through G) are analyzed by 1‐way ANOVA (Tukey post‐test). Data of (D) is analyzed by Mantel‐Cox test. ANP indicates atrial natriuretic peptide; BNP, brain natriuretic peptide; DCM, dilated cardiomyopathy; LVEF, left ventricular ejection fraction; LVFS, left ventricular fractional shortening; LVIDd, left ventricular internal dimensions at end‐diastole; and LVIDs, left ventricular internal dimensions at end‐systole. *P<0.05 between groups; **P<0.01 between groups; ***P<0.005 between groups; and ****P<0.001 between groups. Col indicates colchicine.
Figure 2. Colchicine reduced the recruitment of…
Figure 2. Colchicine reduced the recruitment of neutrophils.
A, ELISA analysis of inflammatory factor interleukin‐1, interleukin‐6, and tumor necrosis factor‐α in murine plasma at the end point of experiment (n=8–12 each group). B, ELISA analysis of GDF15 in murine plasma at the end point of experiment (n=8–12 each group). C, Relative expression of inflammatory cytokine interleukin‐1 mRNA, interleukin‐6 mRNA, and tumor necrosis factor‐α mRNA are normalized to GAPDH mRNA in murine cardiac tissues at the end point of experiment (n=5 each group). D and E, Representative flow cytometric and statistical analysis of CD11b+Ly6G+ neutrophil in the blood (D) and CD45+Ly6G+ neutrophil in the heart € (n=6 each group). F, Representative immunofluorescence images of cardiac tissues staining with Ly6G (green), cTNT (cardiac troponin T) (red), and DAPI (blue). Frequency of Ly6G‐positive neutrophils is shown on the right (n=6–8 each group). Five images per mouse are evaluated. Scale bar=50 μm. Data are shown as mean±SEM. All data statistical significance are determined by 1‐way ANOVA (Tukey post‐test). cTNT indicates cardiac troponin T; DCM, dilated cardiomyopathy; GDF15, growth and differentiation factor 15; IL‐1, interleukin‐1; IL‐6, interleukin‐6; and TNF‐α, tumor necrosis factor‐α. *P<0.05 between groups; **P<0.01 between groups; ***P<0.005 between groups; and ****P<0.001 between groups. Col indicates colchicine; Ly6G, lymphocyte antigen 6 complex locus G6D.
Figure 3. NLRP3 (NOD‐like receptor protein 3)…
Figure 3. NLRP3 (NOD‐like receptor protein 3) inflammasome pathway was inhibited by colchicine treatment.
A and B, Representative immunoblots and the corresponding analysis of NLRP3 inflammasome pathway in cardiac tissues at the end point of experiment. GAPDH shows as loading control. C, Representative immunofluorescence images of cardiac tissues staining with ASC (green), cTNT (cardiac troponin T) (red), and DAPI (blue). Frequency of ASC speck is shown on the right (n=6–8 each group). Five images per mouse are evaluated. Scale bar=50 μm. Data are shown as mean±SEM. All data statistical significance are determined by 1‐way ANOVA (Tukey post‐test). cTNT indicates cardiac troponin T; DCM, dilated cardiomyopathy; IL‐1β, interleukin‐1β; and NLRP3, NOD‐like receptor protein 3. *P<0.05 between groups; **P<0.01 between groups; ***P<0.005 between groups; and ****P<0.001 between groups. AIM2 indicates absent in melanoma 2; ASC, apoptosis‐associated speck‐like protein with CARD domain; Col, colchicine.
Figure 4. SIRT2 (Sirtuin 2) is involved…
Figure 4. SIRT2 (Sirtuin 2) is involved in the anti‐inflammation effect of colchicine.
A, Acetylated NLRP3 (NOD‐like receptor protein 3) levels in the cardiac tissues of mice at the end point of experiment. Immunoglobulin G is the negative control and GAPDH is the input loading control. Data are representative of 3 independent experiments. Western blot analysis on expression of SIRT2 in cardiac tissues at the end of experiment (n=6 each group) on the right. B, Relative expression of SIRT2 mRNA is normalized to GAPDH mRNA in cardiac tissues (n=4 each group). C, Relative expression of inflammatory cytokine interleukin‐1 mRNA and NLRP3 mRNA are normalized to GAPDH mRNA in primary neutrophil that are treated +/− colchicine (10 μmol/L, 2 hours), +/− SIRT2 inhibitor AGK2 (10 μmol/L, 2 hours) or challenged with doxorubicin (5 μmol/L, 2 hours) and inflammasome activators nigericin (10 μmol/L, 2 hours); n=6 each group. D, Western blot analysis on expression of NLRP3 inflammasome pathway in primary neutrophil that are treated +/− colchicine (10 μmol/L, 2 hours), +/− SIRT2 inhibitor AGK2 (10 μmol/L, 2 hours) or challenged with doxorubicin (5 μmol/L, 2 hours) and inflammasome activators nigericin (10 μmol/L, 2 hours). GAPDH shows loading control. Data are representative of 3 independent experiments. Data are shown as mean±SEM. Data of (A and B) are analyzed by 1‐way ANOVA (Tukey post‐test). Data of (C and D) are analyzed by 2‐way ANOVA (Bonferroni post‐test). DCM indicates dilated cardiomyopathy; IL‐1β, interleukin‐1β; NLRP3, NOD‐like receptor protein 3; and SIRT2, Sirtuin 2. *P<0.05 between groups; **P<0.01 between groups; ***P<0.005 between groups; and ****P<0.001 between groups. AGK2 indicates a reversible inhibitor of SIRT2; AIM2, absent in melanoma 2; ASC, apoptosis‐associated speck‐like protein with CARD domain; Col, colchicine; IB, immunoblot; IP, immunoprecipitation; Ly6G, lymphocyte antigen 6 complex locus G6D.
Figure 5. Colchicine promotes the deacetylation of…
Figure 5. Colchicine promotes the deacetylation of NLRP3 (NOD‐like receptor protein 3) by SIRT2 (Sirtuin 2).
A, Representative immunofluorescence images of cardiac tissues staining with NLRP3 (green), SIRT2 (red), and DAPI (blue). (n=6–8 each group). Five images per mouse are evaluated. Scale bar=50 μm. B, Representative immunofluorescence images of primary neutrophils that are treated +/− colchicine (10 μmol/L, 2 hours), +/− SIRT2 inhibitor AGK2 (10 μmol/L, 2 hours) or challenged with doxorubicin (5 μmol/L, 2 hours) and inflammasome activators nigericin (10 μmol/L, 2 hours). NLRP3 (green), SIRT2 (red) and DAPI (blue). Scale bar=20 μm. Data are representative of 3 independent experiments. C, The effect of SIRT2 on the deacetylation of NLRP3 is evaluated by coimmunoprecipitation. NLRP3‐Flag plasmid is transfected into NB4 cell line, then NB4 cell line is treated with +/− colchicine (10 μmol/L, 2 hours), +/− SIRT2 inhibitor AGK2 (10 μmol/L, 2 hours) or challenged with doxorubicin (5 μmol/L, 2 hours) and inflammasome activators nigericin (10 μmol/L, 2 hours). Cell lysates are immunopurified with anti‐Flag antibody. Then the expression of acetylated lysine is measured by IP. GAPDH is the input loading control. Data are representative of 3 independent experiments. DCM indicates dilated cardiomyopathy; IL‐1β, interleukin‐1β; NLRP3, NOD‐like receptor protein 3; and SIRT2, Sirtuin 2. AGK2, a reversible inhibitor of SIRT2; Col, colchicine; IB, immunoblot; IP, Immunoprecipitation.
Figure 6. Inhibition of SIRT2 (Sirtuin 2)…
Figure 6. Inhibition of SIRT2 (Sirtuin 2) abrogates the beneficial effects of colchicine on dilated cardiomyopathy mice.
A through C, Cardiac function is analyzed by 2‐dimensional murine echocardiography, left ventricular (LV) ejection fraction, LV fractional shortening, LV internal dimensions at end‐diastole, and LV internal dimensions at end‐systole values are measured at the end point of experiment (n=10 each group). D, Kaplan–Meier survival curve for each group mice followed for 4 weeks with or without doxorubicin challenge (n=25 each group). E, The changes of body weight from beginning to end (n=10 each group). F, Relative expression of ANP (atrial natriuretic peptide) mRNA and BNP (brain natriuretic peptide) mRNA are normalized to GAPDH mRNA in murine cardiac tissues at the end point of experiment. G and H, The representative hematoxylin and eosin staining (G) and Sirius red staining (H) of hearts in each group are shown. Quantification of interstitial fibrosis (% area) in hearts is shown on the right; n=8 each group. Five images per mouse are evaluated. Scale bar=50 μm. Data are shown as mean±SEM. Data of (B, C, and E through G) are analyzed by 1‐way ANOVA (Tukey post‐test). Data of (D) is analyzed by Mantel‐Cox test. ANP indicates atrial natriuretic peptide; BNP, brain natriuretic peptide; DCM, dilated cardiomyopathy; LVEF, left ventricular ejection fraction; LVFS, left ventricular fractional shortening; LVIDd, left ventricular internal dimensions at end‐diastole; and LVIDs, left ventricular internal dimensions at end‐systole. *P<0.05 between groups; **P<0.01 between groups; ***P<0.005 between groups and ****P<0.001 between groups. AGK2 indicates a reversible inhibitor of SIRT2; Col, colchicine.

References

    1. Japp AG, Gulati A, Cook SA, Cowie MR, Prasad SK. The diagnosis and evaluation of dilated cardiomyopathy. J Am Coll Cardiol. 2016;67:2996–3010. doi: 10.1016/j.jacc.2016.03.590
    1. McNally EM, Mestroni L. Dilated cardiomyopathy: genetic determinants and mechanisms. Circ Res. 2017;121:731–748. doi: 10.1161/CIRCRESAHA.116.309396
    1. Schultheiss HP, Fairweather D, Caforio ALP, Escher F, Hershberger RE, Lipshultz SE, Liu PP, Matsumori A, Mazzanti A, McMurray J, et al. Dilated cardiomyopathy. Nat Rev Dis Primers. 2019;5:32. doi: 10.1038/s41572-019-0084-1
    1. Merlo M, Cannata A, Gobbo M, Stolfo D, Elliott PM, Sinagra G. Evolving concepts in dilated cardiomyopathy. Eur J Heart Fail. 2018;20:228–239. doi: 10.1002/ejhf.1103
    1. Smith ED, Lakdawala NK, Papoutsidakis N, Aubert G, Mazzanti A, McCanta AC, Agarwal PP, Arscott P, Dellefave‐Castillo LM, Vorovich EE, et al. Desmoplakin cardiomyopathy, a fibrotic and inflammatory form of cardiomyopathy distinct from typical dilated or arrhythmogenic right ventricular cardiomyopathy. Circulation. 2020;141:1872–1884. doi: 10.1161/CIRCULATIONAHA.119.044934
    1. Zeng C, Duan F, Hu J, Luo B, Huang B, Lou X, Sun X, Li H, Zhang X, Yin S, et al. NLRP3 inflammasome‐mediated pyroptosis contributes to the pathogenesis of non‐ischemic dilated cardiomyopathy. Redox Biol. 2020;34:101523. doi: 10.1016/j.redox.2020.101523
    1. Caragnano A, Aleksova A, Bulfoni M, Cervellin C, Rolle IG, Veneziano C, Barchiesi A, Mimmi MC, Vascotto C, Finato N, et al. Autophagy and inflammasome activation in dilated cardiomyopathy. J Clin Med. 2019;8:1519. doi: 10.3390/jcm8101519
    1. Tardif JC, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, Pinto FJ, Ibrahim R, Gamra H, Kiwan GS, et al. Efficacy and safety of low‐dose colchicine after myocardial infarction. N Engl J Med. 2019;381:2497–2505. doi: 10.1056/NEJMoa1912388
    1. Imazio M, Nidorf M. Colchicine and the heart. Eur Heart J. 2021;42:2745–2760. doi: 10.1093/eurheartj/ehab221
    1. Slobodnick A, Shah B, Pillinger MH, Krasnokutsky S. Colchicine: old and new. Am J Med. 2015;128:461–470. doi: 10.1016/j.amjmed.2014.12.010
    1. Martinez GJ, Celermajer DS, Patel S. The NLRP3 inflammasome and the emerging role of colchicine to inhibit atherosclerosis‐associated inflammation. Atherosclerosis. 2018;269:262–271. doi: 10.1016/j.atherosclerosis.2017.12.027
    1. He M, Chiang HH, Luo H, Zheng Z, Qiao Q, Wang L, Tan M, Ohkubo R, Mu WC, Zhao S, et al. An acetylation switch of the NLRP3 inflammasome regulates aging‐associated chronic inflammation and insulin resistance. Cell Metab. 2020;31:580–591.e585. doi: 10.1016/j.cmet.2020.01.009
    1. Watroba M, Dudek I, Skoda M, Stangret A, Rzodkiewicz P, Szukiewicz D. Sirtuins, epigenetics and longevity. Ageing Res Rev. 2017;40:11–19. doi: 10.1016/j.arr.2017.08.001
    1. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D'Agostino D, Planavsky N, Lupfer C, Kanneganti TD, et al. The ketone metabolite beta‐hydroxybutyrate blocks NLRP3 inflammasome‐mediated inflammatory disease. Nat Med. 2015;21:263–269. doi: 10.1038/nm.3804
    1. Podyacheva EY, Kushnareva EA, Karpov AA, Toropova YG. Analysis of models of doxorubicin‐induced cardiomyopathy in rats and mice. A modern view from the perspective of the pathophysiologist and the clinician. Front Pharmacol. 2021;12:670479. doi: 10.3389/fphar.2021.670479
    1. Himelman E, Lillo MA, Nouet J, Gonzalez JP, Zhao Q, Xie LH, Li H, Liu T, Wehrens XH, Lampe PD, et al. Prevention of connexin‐43 remodeling protects against duchenne muscular dystrophy cardiomyopathy. J Clin Invest. 2020;130:1713–1727. doi: 10.1172/JCI128190
    1. Li DJ, Sun SJ, Fu JT, Ouyang SX, Zhao QJ, Su L, Ji QX, Sun DY, Zhu JH, Zhang GY, et al. NAD(+)‐boosting therapy alleviates nonalcoholic fatty liver disease via stimulating a novel exerkine Fndc5/irisin. Theranostics. 2021;11:4381–4402. doi: 10.7150/thno.53652
    1. Herster F, Bittner Z, Archer NK, Dickhofer S, Eisel D, Eigenbrod T, Knorpp T, Schneiderhan‐Marra N, Loffler MW, Kalbacher H, et al. Neutrophil extracellular trap‐associated RNA and LL37 enable self‐amplifying inflammation in psoriasis. Nat Commun. 2020;11:105. doi: 10.1038/s41467-019-13756-4
    1. He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, Yang ZH, Zhong CQ, Han J. Gasdermin D is an executor of pyroptosis and required for interleukin‐1beta secretion. Cell Res. 2015;25:1285–1298. doi: 10.1038/cr.2015.139
    1. Zhang C, Chen B, Guo A, Zhu Y, Miller JD, Gao S, Yuan C, Kutschke W, Zimmerman K, Weiss RM, et al. Microtubule‐mediated defects in junctophilin‐2 trafficking contribute to myocyte transverse‐tubule remodeling and Ca2+ handling dysfunction in heart failure. Circulation. 2014;129:1742–1750. doi: 10.1161/CIRCULATIONAHA.113.008452
    1. Misawa T, Takahama M, Kozaki T, Lee H, Zou J, Saitoh T, Akira S. Microtubule‐driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat Immunol. 2013;14:454–460. doi: 10.1038/ni.2550
    1. Li M, Duan L, Cai YL, Li HY, Hao BC, Chen JQ, Liu HB. Growth differentiation factor‐15 is associated with cardiovascular outcomes in patients with coronary artery disease. Cardiovasc Diabetol. 2020;19:120. doi: 10.1186/s12933-020-01092-7
    1. Lok SI, Winkens B, Goldschmeding R, van Geffen AJ, Nous FM, van Kuik J, van der Weide P, Klopping C, Kirkels JH, Lahpor JR, et al. Circulating growth differentiation factor‐15 correlates with myocardial fibrosis in patients with non‐ischaemic dilated cardiomyopathy and decreases rapidly after left ventricular assist device support. Eur J Heart Fail. 2012;14:1249–1256. doi: 10.1093/eurjhf/hfs120
    1. Sun X, Shan A, Wei Z, Xu B. Intravenous mesenchymal stem cell‐derived exosomes ameliorate myocardial inflammation in the dilated cardiomyopathy. Biochem Biophys Res Commun. 2018;503:2611–2618. doi: 10.1016/j.bbrc.2018.08.012
    1. Zhang H, Xu A, Sun X, Yang Y, Zhang L, Bai H, Ben J, Zhu X, Li X, Yang Q, et al. Self‐maintenance of cardiac resident reparative macrophages attenuates doxorubicin‐induced cardiomyopathy through the SR‐A1‐c‐myc axis. Circ Res. 2020;127:610–627. doi: 10.1161/CIRCRESAHA.119.316428
    1. Fernandez‐Ruiz I. Low‐dose colchicine shows promise in chronic coronary disease. Nat Rev Cardiol. 2020;17:680–681. doi: 10.1038/s41569-020-00456-6
    1. Deftereos S, Giannopoulos G, Panagopoulou V, Bouras G, Raisakis K, Kossyvakis C, Karageorgiou S, Papadimitriou C, Vastaki M, Kaoukis A, et al. Anti‐inflammatory treatment with colchicine in stable chronic heart failure: a prospective, randomized study. JACC Heart Fail. 2014;2:131–137. doi: 10.1016/j.jchf.2013.11.006
    1. Wu Q, Liu H, Liao J, Zhao N, Tse G, Han B, Chen L, Huang Z, Du Y. Colchicine prevents atrial fibrillation promotion by inhibiting IL‐1beta‐induced IL‐6 release and atrial fibrosis in the rat sterile pericarditis model. Biomed Pharmacother. 2020;129:110384. doi: 10.1016/j.biopha.2020.110384
    1. Shah B, Pillinger M, Zhong H, Cronstein B, Xia Y, Lorin JD, Smilowitz NR, Feit F, Ratnapala N, Keller NM, et al. Effects of acute colchicine administration prior to percutaneous coronary intervention: COLCHICINE‐PCI randomized trial. Circ Cardiovasc Interv. 2020;13:e008717. doi: 10.1161/CIRCINTERVENTIONS.119.008717
    1. Martini E, Kunderfranco P, Peano C, Carullo P, Cremonesi M, Schorn T, Carriero R, Termanini A, Colombo FS, Jachetti E, et al. Single‐cell sequencing of mouse heart immune infiltrate in pressure overload‐driven heart failure reveals extent of immune activation. Circulation. 2019;140:2089–2107. doi: 10.1161/CIRCULATIONAHA.119.041694
    1. Vaidya K, Tucker B, Kurup R, Khandkar C, Pandzic E, Barraclough J, Machet J, Misra A, Kavurma M, Martinez G, et al. Colchicine inhibits neutrophil extracellular trap formation in patients with acute coronary syndrome after percutaneous coronary intervention. J Am Heart Assoc. 2021;10:e018993. doi: 10.1161/JAHA.120.018993
    1. Xu J, Kimball TR, Lorenz JN, Brown DA, Bauskin AR, Klevitsky R, Hewett TE, Breit SN, Molkentin JD. GDF15/MIC‐1 functions as a protective and antihypertrophic factor released from the myocardium in association with SMAD protein activation. Circ Res. 2006;98:342–350. doi: 10.1161/01.RES.0000202804.84885.d0
    1. May BM, Kochi AN, Magalhaes APA, Scolari F, Zimerman A, Andrades M, Zimerman LI, Rohde LE, Pimentel M. Growth/differentiation factor‐15 (GDF‐15) as a predictor of serious arrhythmic events in patients with nonischemic dilated cardiomyopathy. J Electrocardiol. 2022;70:19–23. doi: 10.1016/j.jelectrocard.2021.10.002
    1. Weng JH, Koch PD, Luan HH, Tu HC, Shimada K, Ngan I, Ventura R, Jiang R, Mitchison TJ. Colchicine acts selectively in the liver to induce hepatokines that inhibit myeloid cell activation. Nat Metab. 2021;3:513–522. doi: 10.1038/s42255-021-00366-y
    1. Wei S, Ma W, Zhang B, Li W. NLRP3 inflammasome: a promising therapeutic target for drug‐induced toxicity. Front Cell Dev Biol. 2021;9:634607. doi: 10.3389/fcell.2021.634607
    1. Leung YY, Yao Hui LL, Kraus VB. Colchicine—update on mechanisms of action and therapeutic uses. Semin Arthritis Rheum. 2015;45:341–350. doi: 10.1016/j.semarthrit.2015.06.013
    1. Wang Y, Viollet B, Terkeltaub R, Liu‐Bryan R. AMP‐activated protein kinase suppresses urate crystal‐induced inflammation and transduces colchicine effects in macrophages. Ann Rheum Dis. 2016;75:286–294. doi: 10.1136/annrheumdis-2014-206074
    1. Dubey KK, Kumar P, Labrou NE, Shukla P. Biotherapeutic potential and mechanisms of action of colchicine. Crit Rev Biotechnol. 2017;37:1038–1047. doi: 10.1080/07388551.2017.1303804
    1. Matsushima S, Sadoshima J. The role of sirtuins in cardiac disease. Am J Physiol Heart Circ Physiol. 2015;309:H1375–H1389. doi: 10.1152/ajpheart.00053.2015
    1. Singh CK, Chhabra G, Ndiaye MA, Garcia‐Peterson LM, Mack NJ, Ahmad N. The role of sirtuins in antioxidant and redox signaling. Antioxid Redox Signal. 2018;28:643–661. doi: 10.1089/ars.2017.7290
    1. Zhao L, Qi Y, Xu L, Tao X, Han X, Yin L, Peng J. MicroRNA‐140‐5p aggravates doxorubicin‐induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2. Redox Biol. 2018;15:284–296. doi: 10.1016/j.redox.2017.12.013
    1. Tang X, Chen XF, Wang NY, Wang XM, Liang ST, Zheng W, Lu YB, Zhao X, Hao DL, Zhang ZQ, et al. SIRT2 acts as a cardioprotective deacetylase in pathological cardiac hypertrophy. Circulation. 2017;136:2051–2067. doi: 10.1161/CIRCULATIONAHA.117.028728
    1. Yuan Q, Zhan L, Zhou QY, Zhang LL, Chen XM, Hu XM, Yuan XC. SIRT2 regulates microtubule stabilization in diabetic cardiomyopathy. Eur J Pharmacol. 2015;764:554–561. doi: 10.1016/j.ejphar.2015.07.045
    1. Kida Y, Goligorsky MS. Sirtuins, cell senescence, and vascular aging. Can J Cardiol. 2016;32:634–641. doi: 10.1016/j.cjca.2015.11.022
    1. Watanabe H, Inaba Y, Kimura K, Matsumoto M, Kaneko S, Kasuga M, Inoue H. Sirt2 facilitates hepatic glucose uptake by deacetylating glucokinase regulatory protein. Nat Commun. 2018;9:30. doi: 10.1038/s41467-017-02537-6
    1. Krishnan J, Danzer C, Simka T, Ukropec J, Walter KM, Kumpf S, Mirtschink P, Ukropcova B, Gasperikova D, Pedrazzini T, et al. Dietary obesity‐associated Hif1α activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2‐NAD+ system. Genes Dev. 2012;26:259–270. doi: 10.1101/gad.180406.111
    1. Caporizzo MA, Chen CY, Prosser BL. Cardiac microtubules in health and heart disease. Exp Biol Med. 2019;244:1255–1272. doi: 10.1177/1535370219868960
    1. Kerfant BG, Vassort G, Gomez AM. Microtubule disruption by colchicine reversibly enhances calcium signaling in intact rat cardiac myocytes. Circ Res. 2001;88:E59–E65. doi: 10.1161/hh0701.090462
    1. Lu YY, Chen YC, Kao YH, Lin YK, Yeh YH, Chen SA, Chen YJ. Colchicine modulates calcium homeostasis and electrical property of HL‐1 cells. J Cell Mol Med. 2016;20:1182–1190. doi: 10.1111/jcmm.12818
    1. Caporizzo MA, Chen CY, Bedi K, Margulies KB, Prosser BL. Microtubules increase diastolic stiffness in failing human cardiomyocytes and myocardium. Circulation. 2020;141:902–915. doi: 10.1161/CIRCULATIONAHA.119.043930
    1. El‐Sayed EM, Mansour AM, El‐Sawy WS. Protective effect of proanthocyanidins against doxorubicin‐induced nephrotoxicity in rats. J Biochem Mol Toxicol. 2017;31:e21965. doi: 10.1002/jbt.21965
    1. Jacevic V, Djordjevic A, Srdjenovic B, Milic‐Tores V, Segrt Z, Dragojevic‐Simic V, Kuca K. Fullerenol nanoparticles prevents doxorubicin‐induced acute hepatotoxicity in rats. Exp Mol Pathol. 2017;102:360–369. doi: 10.1016/j.yexmp.2017.03.005
    1. Tangpong J, Cole MP, Sultana R, Estus S, Vore M, St Clair W, Ratanachaiyavong S, St Clair DK, Butterfield DA. Adriamycin‐mediated nitration of manganese superoxide dismutase in the central nervous system: insight into the mechanism of chemobrain. J Neurochem. 2007;100:191–201. doi: 10.1111/j.1471-4159.2006.04179.x
    1. Kim S, Jung ES, Lee J, Heo NJ, Na KY, Han JS. Effects of colchicine on renal fibrosis and apoptosis in obstructed kidneys. Korean J Intern Med. 2018;33:568–576. doi: 10.3904/kjim.2016.131
    1. Qu ZA, Ma XJ, Huang SB, Hao XR, Li DM, Feng KY, Wang WM. SIRT2 inhibits oxidative stress and inflammatory response in diabetic osteoarthritis. Eur Rev Med Pharmacol Sci. 2020;24:2855–2864. doi: 10.26355/eurrev_202003_20649
    1. Ahmed DS, Isnard S, Lin J, Routy B, Routy JP. GDF15/GFRAL pathway as a metabolic signature for cachexia in patients with cancer. J Cancer. 2021;12:1125–1132. doi: 10.7150/jca.50376
    1. Johnen H, Lin S, Kuffner T, Brown DA, Tsai VW, Bauskin AR, Wu L, Pankhurst G, Jiang L, Junankar S, et al. Tumor‐induced anorexia and weight loss are mediated by the TGF‐beta superfamily cytokine MIC‐1. Nat Med. 2007;13:1333–1340. doi: 10.1038/nm1677
    1. Kempf T, Zarbock A, Widera C, Butz S, Stadtmann A, Rossaint J, Bolomini‐Vittori M, Korf‐Klingebiel M, Napp LC, Hansen B, et al. GDF‐15 is an inhibitor of leukocyte integrin activation required for survival after myocardial infarction in mice. Nat Med. 2011;17:581–588. doi: 10.1038/nm.2354

Source: PubMed

3
Abonneren