Protective Effects of Curcumin in Cardiovascular Diseases—Impact on Oxidative Stress and Mitochondria
Abstract
:1. Introduction
1.1. Cardiovascular Diseases
1.2. Curcumin as Nutraceutical with Pleiotropic Actions
2. Cardiovascular Diseases and Risk Factors
2.1. Atherosclerosis and Myocardial Infarction
2.2. Brown and Perivascular Adipose Tissue in Cardiovascular Diseases
2.3. Aging as a Non-Modifiable Risk Factor for Cardiovascular Diseases
2.4. Obesity as a Modifiable Risk Factor for Cardiovascular Diseases
3. Protective Role of Curcumin in Cardiovascular Diseases
3.1. Effects of Curcumin on Cellular Senescence and Age-Related Cardiovascular Dysfunction
3.2. Effects of Curcumin on Adipose Tissue and Obesity
3.3. Protective Effects of Curcumin in Atherosclerosis
3.4. Protective Effects of Curcumin in Myocardial Infarction
4. Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- Mathers, C.D.; Loncar, D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006, 3, e442. [Google Scholar] [CrossRef] [Green Version]
- World Health Organization. Global Health Risks: Mortality and Burden of Disease Attributable to Selected Major Risks; World Health Organization: Geneva, Switzerland, 2009. [Google Scholar]
- Vogel, A.; Pelletier, J. Examen chimique de la racine de Curcuma. J. De Pharm. 1815, I, 289–300. [Google Scholar]
- Vogel, A. Curcumin. Justus Liebigs Ann. Chem. 1842, 44, 297–298. [Google Scholar]
- Miłobȩdzka, J.; Kostanecki, S.V.; Lampe, V. Zur Kenntnis des Curcumins. Ber. Dtsch. Chem. Ges. 1910, 43, 2163–2170. [Google Scholar] [CrossRef] [Green Version]
- Lampe, V. Synthese von Curcumin. Ber. Dtsch. Chem. Ges. 1918, 51, 1347–1355. [Google Scholar] [CrossRef] [Green Version]
- Schraufstätter, E.; Bernt, H. Antibacterial action of curcumin and related compounds. Nature 1949, 164, 456–457. [Google Scholar] [CrossRef] [PubMed]
- Srimal, R.C.; Dhawan, B.N. Pharmacology of diferuloyl methane (curcumin), a non-steroidal anti-inflammatory agent. J. Pharm. Pharmacol. 1973, 25, 447–452. [Google Scholar] [CrossRef] [PubMed]
- Sharma, O.P. Antioxidant activity of curcumin and related compounds. Biochem. Pharmacol. 1976, 25, 1811–1812. [Google Scholar] [CrossRef]
- Kuttan, R.; Bhanumathy, P.; Nirmala, K.; George, M.C. Potential anticancer activity of turmeric (Curcuma longa). Cancer Lett. 1985, 29, 197–202. [Google Scholar] [CrossRef]
- Gupta, S.C.; Patchva, S.; Aggarwal, B.B. Therapeutic roles of curcumin: Lessons learned from clinical trials. AAPS J. 2013, 15, 195–218. [Google Scholar] [CrossRef] [Green Version]
- Gimbrone, M.A., Jr.; García-Cardeña, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef] [Green Version]
- Förstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef] [PubMed]
- Linton, M.F.; Yancey, P.G.; Davies, S.S.; Jerome, W.G.; Linton, E.F.; Song, W.L.; Doran, A.C.; Vickers, K.C. The Role of Lipids and Lipoproteins in Atherosclerosis. In Endotext; Feingold, K.R., Anawalt, B., Boyce, A., Chrousos, G., de Herder, W.W., Dhatariya, K., Dungan, K., Hershman, J.M., Hofland, J., Kalra, S., et al., Eds.; © 2000–2021; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
- Leonarduzzi, G.; Chiarpotto, E.; Biasi, F.; Poli, G. 4-Hydroxynonenal and cholesterol oxidation products in atherosclerosis. Mol. Nutr. Food Res. 2005, 49, 1044–1049. [Google Scholar] [CrossRef]
- Ahotupa, M. Oxidized lipoprotein lipids and atherosclerosis. Free Radic. Res. 2017, 51, 439–447. [Google Scholar] [CrossRef]
- Oliveira, H.C.; Cosso, R.G.; Alberici, L.C.; Maciel, E.N.; Salerno, A.G.; Dorighello, G.G.; Velho, J.A.; de Faria, E.C.; Vercesi, A.E. Oxidative stress in atherosclerosis-prone mouse is due to low antioxidant capacity of mitochondria. FASEB J. 2005, 19, 278–280. [Google Scholar] [CrossRef] [PubMed]
- Harrison, C.M.; Pompilius, M.; Pinkerton, K.E.; Ballinger, S.W. Mitochondrial oxidative stress significantly influences atherogenic risk and cytokine-induced oxidant production. Environ. Health Perspect. 2011, 119, 676–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Primers 2019, 5, 56. [Google Scholar] [CrossRef] [PubMed]
- Chouchani, E.T.; Pell, V.R.; James, A.M.; Work, L.M.; Saeb-Parsy, K.; Frezza, C.; Krieg, T.; Murphy, M.P. A Unifying Mechanism for Mitochondrial Superoxide Production during Ischemia-Reperfusion Injury. Cell Metab. 2016, 23, 254–263. [Google Scholar] [CrossRef] [Green Version]
- Turner, N.A.; Porter, K.E. Function and fate of myofibroblasts after myocardial infarction. Fibrogenes. Tissue Repair 2013, 6, 5. [Google Scholar] [CrossRef] [Green Version]
- Miquerol, L.; Thireau, J.; Bideaux, P.; Sturny, R.; Richard, S.; Kelly, R.G. Endothelial plasticity drives arterial remodeling within the endocardium after myocardial infarction. Circ. Res. 2015, 116, 1765–1771. [Google Scholar] [CrossRef] [Green Version]
- Gonnissen, S.; Ptok, J.; Goy, C.; Jander, K.; Jakobs, P.; Eckermann, O.; Kaisers, W.; von Ameln, F.; Timm, J.; Ale-Agha, N.; et al. High Concentration of Low-Density Lipoprotein Results in Disturbances in Mitochondrial Transcription and Functionality in Endothelial Cells. Oxid. Med. Cell. Longev. 2019, 2019, 7976382. [Google Scholar] [CrossRef]
- Ale-Agha, N.; Jakobs, P.; Goy, C.; Zurek, M.; Rosen, J.; Dyballa-Rukes, N.; Metzger, S.; Greulich, J.; von Ameln, F.; Eckermann, O.; et al. Mitochondrial Telomerase Reverse Transcriptase Protects from Myocardial Ischemia/reperfusion Injury by Improving Complex I Composition and Function. Circulation 2021, 144, 1876–1890. [Google Scholar] [CrossRef] [PubMed]
- Gesta, S.; Tseng, Y.H.; Kahn, C.R. Developmental origin of fat: Tracking obesity to its source. Cell 2007, 131, 242–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nedergaard, J.; Bengtsson, T.; Cannon, B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 2007, 293, E444–E452. [Google Scholar] [CrossRef]
- Virtanen, K.A.; Lidell, M.E.; Orava, J.; Heglind, M.; Westergren, R.; Niemi, T.; Taittonen, M.; Laine, J.; Savisto, N.J.; Enerbäck, S.; et al. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. 2009, 360, 1518–1525. [Google Scholar] [CrossRef] [PubMed]
- Zingaretti, M.C.; Crosta, F.; Vitali, A.; Guerrieri, M.; Frontini, A.; Cannon, B.; Nedergaard, J.; Cinti, S. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J. 2009, 23, 3113–3120. [Google Scholar] [CrossRef]
- Thyagarajan, B.; Foster, M.T. Beiging of white adipose tissue as a therapeutic strategy for weight loss in humans. Horm. Mol. Biol. Clin. Investig. 2017, 31, 20170016. [Google Scholar] [CrossRef]
- Bartelt, A.; Bruns, O.T.; Reimer, R.; Hohenberg, H.; Ittrich, H.; Peldschus, K.; Kaul, M.G.; Tromsdorf, U.I.; Weller, H.; Waurisch, C.; et al. Brown adipose tissue activity controls triglyceride clearance. Nat. Med. 2011, 17, 200–205. [Google Scholar] [CrossRef]
- Geerling, J.J.; Boon, M.R.; van der Zon, G.C.; van den Berg, S.A.; van den Hoek, A.M.; Lombès, M.; Princen, H.M.; Havekes, L.M.; Rensen, P.C.; Guigas, B. Metformin lowers plasma triglycerides by promoting VLDL-triglyceride clearance by brown adipose tissue in mice. Diabetes 2014, 63, 880–891. [Google Scholar] [CrossRef] [Green Version]
- Berbée, J.F.; Boon, M.R.; Khedoe, P.P.; Bartelt, A.; Schlein, C.; Worthmann, A.; Kooijman, S.; Hoeke, G.; Mol, I.M.; John, C.; et al. Brown fat activation reduces hypercholesterolaemia and protects from atherosclerosis development. Nat. Commun. 2015, 6, 6356. [Google Scholar] [CrossRef] [Green Version]
- Raiko, J.; Orava, J.; Savisto, N.; Virtanen, K.A. High Brown Fat Activity Correlates With Cardiovascular Risk Factor Levels Cross-Sectionally and Subclinical Atherosclerosis at 5-Year Follow-Up. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1289–1295. [Google Scholar] [CrossRef]
- Becher, T.; Palanisamy, S.; Kramer, D.J.; Eljalby, M.; Marx, S.J.; Wibmer, A.G.; Butler, S.D.; Jiang, C.S.; Vaughan, R.; Schöder, H.; et al. Brown adipose tissue is associated with cardiometabolic health. Nat. Med. 2021, 27, 58–65. [Google Scholar] [CrossRef]
- Hildebrand, S.; Stümer, J.; Pfeifer, A. PVAT and Its Relation to Brown, Beige, and White Adipose Tissue in Development and Function. Front. Physiol. 2018, 9, 70. [Google Scholar] [CrossRef] [Green Version]
- Fitzgibbons, T.P.; Kogan, S.; Aouadi, M.; Hendricks, G.M.; Straubhaar, J.; Czech, M.P. Similarity of mouse perivascular and brown adipose tissues and their resistance to diet-induced inflammation. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1425–H1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padilla, J.; Jenkins, N.T.; Vieira-Potter, V.J.; Laughlin, M.H. Divergent phenotype of rat thoracic and abdominal perivascular adipose tissues. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2013, 304, R543–R552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Victorio, J.A.; Fontes, M.T.; Rossoni, L.V.; Davel, A.P. Different Anti-Contractile Function and Nitric Oxide Production of Thoracic and Abdominal Perivascular Adipose Tissues. Front. Physiol. 2016, 7, 295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qi, X.Y.; Qu, S.L.; Xiong, W.H.; Rom, O.; Chang, L.; Jiang, Z.S. Perivascular adipose tissue (PVAT) in atherosclerosis: A double-edged sword. Cardiovasc. Diabetol. 2018, 17, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, L.; Villacorta, L.; Li, R.; Hamblin, M.; Xu, W.; Dou, C.; Zhang, J.; Wu, J.; Zeng, R.; Chen, Y.E. Loss of perivascular adipose tissue on peroxisome proliferator-activated receptor-γ deletion in smooth muscle cells impairs intravascular thermoregulation and enhances atherosclerosis. Circulation 2012, 126, 1067–1078. [Google Scholar] [CrossRef] [PubMed]
- Baltieri, N.; Guizoni, D.M.; Victorio, J.A.; Davel, A.P. Protective Role of Perivascular Adipose Tissue in Endothelial Dysfunction and Insulin-Induced Vasodilatation of Hypercholesterolemic LDL Receptor-Deficient Mice. Front. Physiol. 2018, 9, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xia, N.; Horke, S.; Habermeier, A.; Closs, E.I.; Reifenberg, G.; Gericke, A.; Mikhed, Y.; Münzel, T.; Daiber, A.; Förstermann, U.; et al. Uncoupling of Endothelial Nitric Oxide Synthase in Perivascular Adipose Tissue of Diet-Induced Obese Mice. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 78–85. [Google Scholar] [CrossRef] [Green Version]
- Bar, A.; Kieronska-Rudek, A.; Proniewski, B.; Suraj-Prażmowska, J.; Czamara, K.; Marczyk, B.; Matyjaszczyk-Gwarda, K.; Jasztal, A.; Kuś, E.; Majka, Z.; et al. In Vivo Magnetic Resonance Imaging-Based Detection of Heterogeneous Endothelial Response in Thoracic and Abdominal Aorta to Short-Term High-Fat Diet Ascribed to Differences in Perivascular Adipose Tissue in Mice. J. Am. Heart Assoc. 2020, 9, e016929. [Google Scholar] [CrossRef]
- Czamara, K.; Majka, Z.; Sternak, M.; Koziol, M.; Kostogrys, R.B.; Chlopicki, S.; Kaczor, A. Distinct Chemical Changes in Abdominal but Not in Thoracic Aorta upon Atherosclerosis Studied Using Fiber Optic Raman Spectroscopy. Int. J. Mol. Sci. 2020, 21, 4838. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Wang, Z.; Wang, C.; Ma, Q.; Zhao, Y. Perivascular adipose tissue-derived adiponectin inhibits collar-induced carotid atherosclerosis by promoting macrophage autophagy. PLoS ONE 2015, 10, e0124031. [Google Scholar]
- Sena, C.M.; Pereira, A.; Fernandes, R.; Letra, L.; Seiça, R.M. Adiponectin improves endothelial function in mesenteric arteries of rats fed a high-fat diet: Role of perivascular adipose tissue. Br. J. Pharmacol. 2017, 174, 3514–3526. [Google Scholar] [CrossRef] [Green Version]
- Fernández-Friera, L.; Peñalvo, J.L.; Fernández-Ortiz, A.; Ibañez, B.; López-Melgar, B.; Laclaustra, M.; Oliva, B.; Mocoroa, A.; Mendiguren, J.; Martínez de Vega, V.; et al. Prevalence, Vascular Distribution, and Multiterritorial Extent of Subclinical Atherosclerosis in a Middle-Aged Cohort: The PESA (Progression of Early Subclinical Atherosclerosis) Study. Circulation 2015, 131, 2104–2113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Virani, S.S.; Alonso, A.; Aparicio, H.J.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics-2021 Update: A Report from the American Heart Association. Circulation 2021, 143, e254–e743. [Google Scholar] [CrossRef] [PubMed]
- Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update From the GBD 2019 Study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef] [PubMed]
- Driver, J.A.; Djoussé, L.; Logroscino, G.; Gaziano, J.M.; Kurth, T. Incidence of cardiovascular disease and cancer in advanced age: Prospective cohort study. BMJ 2008, 337, a2467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodgers, J.L.; Jones, J.; Bolleddu, S.I.; Vanthenapalli, S.; Rodgers, L.E.; Shah, K.; Karia, K.; Panguluri, S.K. Cardiovascular Risks Associated with Gender and Aging. J. Cardiovasc. Dev. Dis. 2019, 6, 19. [Google Scholar] [CrossRef] [Green Version]
- Paneni, F.; Diaz Cañestro, C.; Libby, P.; Lüscher, T.F.; Camici, G.G. The Aging Cardiovascular System: Understanding It at the Cellular and Clinical Levels. J. Am. Coll. Cardiol. 2017, 69, 1952–1967. [Google Scholar] [CrossRef]
- Pan, X.X.; Ruan, C.C.; Liu, X.Y.; Kong, L.R.; Ma, Y.; Wu, Q.H.; Li, H.Q.; Sun, Y.J.; Chen, A.Q.; Zhao, Q.; et al. Perivascular adipose tissue-derived stromal cells contribute to vascular remodeling during aging. Aging Cell. 2019, 18, e12969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mijit, M.; Caracciolo, V.; Melillo, A.; Amicarelli, F.; Giordano, A. Role of p53 in the Regulation of Cellular Senescence. Biomolecules 2020, 10, 420. [Google Scholar] [CrossRef] [Green Version]
- Katsuumi, G.; Shimizu, I.; Yoshida, Y.; Minamino, T. Vascular Senescence in Cardiovascular and Metabolic Diseases. Front. Cardiovasc. Med. 2018, 5, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hernansanz-Agustín, P.; Enríquez, J.A. Generation of Reactive Oxygen Species by Mitochondria. Antioxidants 2021, 10, 415. [Google Scholar] [CrossRef] [PubMed]
- Rizvi, F.; Preston, C.C.; Emelyanova, L.; Yousufuddin, M.; Viqar, M.; Dakwar, O.; Ross, G.R.; Faustino, R.S.; Holmuhamedov, E.L.; Jahangir, A. Effects of Aging on Cardiac Oxidative Stress and Transcriptional Changes in Pathways of Reactive Oxygen Species Generation and Clearance. J. Am. Heart Assoc. 2021, 10, e019948. [Google Scholar] [CrossRef] [PubMed]
- Ago, T.; Matsushima, S.; Kuroda, J.; Zablocki, D.; Kitazono, T.; Sadoshima, J. The NADPH oxidase Nox4 and aging in the heart. Aging 2010, 2, 1012–1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Canugovi, C.; Stevenson, M.D.; Vendrov, A.E.; Hayami, T.; Robidoux, J.; Xiao, H.; Zhang, Y.Y.; Eitzman, D.T.; Runge, M.S.; Madamanchi, N.R. Increased mitochondrial NADPH oxidase 4 (NOX4) expression in aging is a causative factor in aortic stiffening. Redox Biol. 2019, 26, 101288. [Google Scholar] [CrossRef]
- Ikeda, Y.; Sciarretta, S.; Nagarajan, N.; Rubattu, S.; Volpe, M.; Frati, G.; Sadoshima, J. New insights into the role of mitochondrial dynamics and autophagy during oxidative stress and aging in the heart. Oxid. Med. Cell. Longev. 2014, 2014, 210934. [Google Scholar] [CrossRef] [PubMed]
- Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxid. Redox Signal 2018, 29, 1727–1745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vomund, S.; Schäfer, A.; Parnham, M.J.; Brüne, B.; von Knethen, A. Nrf2, the Master Regulator of Anti-Oxidative Responses. Int. J. Mol. Sci. 2017, 18, 2772. [Google Scholar] [CrossRef] [Green Version]
- Holmström, K.M.; Baird, L.; Zhang, Y.; Hargreaves, I.; Chalasani, A.; Land, J.M.; Stanyer, L.; Yamamoto, M.; Dinkova-Kostova, A.T.; Abramov, A.Y. Nrf2 impacts cellular bioenergetics by controlling substrate availability for mitochondrial respiration. Biol. Open 2013, 2, 761–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suh, J.H.; Shenvi, S.V.; Dixon, B.M.; Liu, H.; Jaiswal, A.K.; Liu, R.M.; Hagen, T.M. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc. Natl. Acad. Sci. USA 2004, 101, 3381–3386. [Google Scholar] [CrossRef] [Green Version]
- Shih, P.H.; Yen, G.C. Differential expressions of antioxidant status in aging rats: The role of transcriptional factor Nrf2 and MAPK signaling pathway. Biogerontology 2007, 8, 71–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ungvari, Z.; Bailey-Downs, L.; Sosnowska, D.; Gautam, T.; Koncz, P.; Losonczy, G.; Ballabh, P.; de Cabo, R.; Sonntag, W.E.; Csiszar, A. Vascular oxidative stress in aging: A homeostatic failure due to dysregulation of NRF2-mediated antioxidant response. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H363–H372. [Google Scholar] [CrossRef] [Green Version]
- Gounder, S.S.; Kannan, S.; Devadoss, D.; Miller, C.J.; Whitehead, K.J.; Odelberg, S.J.; Firpo, M.A.; Paine, R., 3rd; Hoidal, J.R.; Abel, E.D.; et al. Impaired transcriptional activity of Nrf2 in age-related myocardial oxidative stress is reversible by moderate exercise training. PLoS ONE 2012, 7, e45697. [Google Scholar] [CrossRef]
- Fulop, G.A.; Kiss, T.; Tarantini, S.; Balasubramanian, P.; Yabluchanskiy, A.; Farkas, E.; Bari, F.; Ungvari, Z.; Csiszar, A. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. Geroscience 2018, 40, 513–521. [Google Scholar] [CrossRef]
- Zhou, T.; Zhang, M.; Zhao, L.; Li, A.; Qin, X. Activation of Nrf2 contributes to the protective effect of Exendin-4 against angiotensin II-induced vascular smooth muscle cell senescence. Am. J. Physiol. Cell Physiol. 2016, 311, C572–C582. [Google Scholar] [CrossRef] [Green Version]
- Wang, R.Y.; Liu, L.H.; Liu, H.; Wu, K.F.; An, J.; Wang, Q.; Liu, Y.; Bai, L.J.; Qi, B.M.; Qi, B.L.; et al. Nrf2 protects against diabetic dysfunction of endothelial progenitor cells via regulating cell senescence. Int. J. Mol. Med. 2018, 42, 1327–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: A pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet 2017, 390, 2627–2642. [Google Scholar] [CrossRef] [Green Version]
- Reilly, J.J.; El-Hamdouchi, A.; Diouf, A.; Monyeki, A.; Somda, S.A. Determining the worldwide prevalence of obesity. Lancet 2018, 391, 1773–1774. [Google Scholar] [CrossRef]
- Astrup, A.; Finer, N. Redefining type 2 diabetes: ‘diabesity’ or ‘obesity dependent diabetes mellitus’? Obes. Rev. 2000, 1, 57–59. [Google Scholar] [CrossRef] [PubMed]
- Després, J.P.; Lemieux, I. Abdominal obesity and metabolic syndrome. Nature 2006, 444, 881–887. [Google Scholar] [CrossRef]
- Vergès, B. Lipid modification in type 2 diabetes: The role of LDL and HDL. Fundam. Clin. Pharmacol. 2009, 23, 681–685. [Google Scholar] [CrossRef] [PubMed]
- Feingold, K.R. Obesity and Dyslipidemia. In Endotext; Feingold, K.R., Anawalt, B., Boyce, A., Chrousos, G., de Herder, W.W., Dhatariya, K., Dungan, K., Hershman, J.M., Hofland, J., Kalra, S., et al., Eds.; © 2000–2021; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
- Leitner, B.P.; Huang, S.; Brychta, R.J.; Duckworth, C.J.; Baskin, A.S.; McGehee, S.; Tal, I.; Dieckmann, W.; Gupta, G.; Kolodny, G.M.; et al. Mapping of human brown adipose tissue in lean and obese young men. Proc. Natl. Acad. Sci. USA 2017, 114, 8649–8654. [Google Scholar] [CrossRef] [Green Version]
- Alcalá, M.; Calderon-Dominguez, M.; Bustos, E.; Ramos, P.; Casals, N.; Serra, D.; Viana, M.; Herrero, L. Increased inflammation, oxidative stress and mitochondrial respiration in brown adipose tissue from obese mice. Sci. Rep. 2017, 7, 16082. [Google Scholar] [CrossRef] [Green Version]
- Aghamohammadzadeh, R.; Unwin, R.D.; Greenstein, A.S.; Heagerty, A.M. Effects of Obesity on Perivascular Adipose Tissue Vasorelaxant Function: Nitric Oxide, Inflammation and Elevated Systemic Blood Pressure. J. Vasc. Res. 2015, 52, 299–305. [Google Scholar] [CrossRef]
- Greenstein, A.S.; Khavandi, K.; Withers, S.B.; Sonoyama, K.; Clancy, O.; Jeziorska, M.; Laing, I.; Yates, A.P.; Pemberton, P.W.; Malik, R.A.; et al. Local inflammation and hypoxia abolish the protective anticontractile properties of perivascular fat in obese patients. Circulation 2009, 119, 1661–1670. [Google Scholar] [CrossRef] [Green Version]
- Saxton, S.N.; Whitley, A.S.; Potter, R.J.; Withers, S.B.; Grencis, R.; Heagerty, A.M. Interleukin-33 rescues perivascular adipose tissue anticontractile function in obesity. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H1387–H1397. [Google Scholar] [CrossRef]
- Aghamohammadzadeh, R.; Greenstein, A.S.; Yadav, R.; Jeziorska, M.; Hama, S.; Soltani, F.; Pemberton, P.W.; Ammori, B.; Malik, R.A.; Soran, H.; et al. Effects of bariatric surgery on human small artery function: Evidence for reduction in perivascular adipocyte inflammation, and the restoration of normal anticontractile activity despite persistent obesity. J. Am. Coll. Cardiol. 2013, 62, 128–135. [Google Scholar] [CrossRef]
- Goldstein, B.J. Insulin resistance: From benign to type 2 diabetes mellitus. Rev. Cardiovasc. Med. 2003, 4 (Suppl. 6), S3–S10. [Google Scholar] [PubMed]
- Gallagher, E.J.; Leroith, D.; Karnieli, E. Insulin resistance in obesity as the underlying cause for the metabolic syndrome. Mt. Sinai J. Med. 2010, 77, 511–523. [Google Scholar] [CrossRef]
- Esser, N.; Legrand-Poels, S.; Piette, J.; Scheen, A.J.; Paquot, N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res. Clin. Pract. 2014, 105, 141–150. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, F.M.; Weschenfelder, J.; Sander, C.; Minkwitz, J.; Thormann, J.; Chittka, T.; Mergl, R.; Kirkby, K.C.; Faßhauer, M.; Stumvoll, M.; et al. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS ONE 2015, 10, e0121971. [Google Scholar] [CrossRef] [PubMed]
- Fain, J.N. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam. Horm. 2006, 74, 443–477. [Google Scholar]
- Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Investig. 2003, 112, 1821–1830. [Google Scholar] [CrossRef] [PubMed]
- Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef]
- Cinti, S.; Mitchell, G.; Barbatelli, G.; Murano, I.; Ceresi, E.; Faloia, E.; Wang, S.; Fortier, M.; Greenberg, A.S.; Obin, M.S. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 2005, 46, 2347–2355. [Google Scholar] [CrossRef] [Green Version]
- Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Investig. 2007, 117, 175–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lumeng, C.N.; DelProposto, J.B.; Westcott, D.J.; Saltiel, A.R. Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 2008, 57, 3239–3246. [Google Scholar] [CrossRef] [Green Version]
- Ricardo-Gonzalez, R.R.; Red Eagle, A.; Odegaard, J.I.; Jouihan, H.; Morel, C.R.; Heredia, J.E.; Mukundan, L.; Wu, D.; Locksley, R.M.; Chawla, A. IL-4/STAT6 immune axis regulates peripheral nutrient metabolism and insulin sensitivity. Proc. Natl. Acad. Sci. USA 2010, 107, 22617–22622. [Google Scholar] [CrossRef] [Green Version]
- Marseglia, L.; Manti, S.; D’Angelo, G.; Nicotera, A.; Parisi, E.; Di Rosa, G.; Gitto, E.; Arrigo, T. Oxidative stress in obesity: A critical component in human diseases. Int. J. Mol. Sci. 2014, 16, 378–400. [Google Scholar] [CrossRef] [Green Version]
- Chrysohoou, C.; Panagiotakos, D.B.; Pitsavos, C.; Skoumas, I.; Papademetriou, L.; Economou, M.; Stefanadis, C. The implication of obesity on total antioxidant capacity in apparently healthy men and women: The ATTICA study. Nutr. Metab. Cardiovasc. Dis. 2007, 17, 590–597. [Google Scholar] [CrossRef]
- Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
- Talior, I.; Yarkoni, M.; Bashan, N.; Eldar-Finkelman, H. Increased glucose uptake promotes oxidative stress and PKC-delta activation in adipocytes of obese, insulin-resistant mice. Am. J. Physiol. Endocrinol. Metab. 2003, 285, E295–E302. [Google Scholar] [CrossRef] [Green Version]
- Houstis, N.; Rosen, E.D.; Lander, E.S. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 2006, 440, 944–948. [Google Scholar] [CrossRef] [PubMed]
- Shin, S.; Wakabayashi, J.; Yates, M.S.; Wakabayashi, N.; Dolan, P.M.; Aja, S.; Liby, K.T.; Sporn, M.B.; Yamamoto, M.; Kensler, T.W. Role of Nrf2 in prevention of high-fat diet-induced obesity by synthetic triterpenoid CDDO-imidazolide. Eur. J. Pharmacol. 2009, 620, 138–144. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.Y.; Kang, B.; Suh, H.J.; Choi, H.S. Parthenolide, a feverfew-derived phytochemical, ameliorates obesity and obesity-induced inflammatory responses via the Nrf2/Keap1 pathway. Pharmacol. Res. 2019, 145, 104259. [Google Scholar] [CrossRef] [PubMed]
- Sampath, C.; Rashid, M.R.; Sang, S.; Ahmedna, M. Green tea epigallocatechin 3-gallate alleviates hyperglycemia and reduces advanced glycation end products via nrf2 pathway in mice with high fat diet-induced obesity. Biomed. Pharmacother. 2017, 87, 73–81. [Google Scholar] [CrossRef] [PubMed]
- Yu, Z.; Shao, W.; Chiang, Y.; Foltz, W.; Zhang, Z.; Ling, W.; Fantus, I.G.; Jin, T. Oltipraz upregulates the nuclear factor (erythroid-derived 2)-like 2 [corrected](NRF2) antioxidant system and prevents insulin resistance and obesity induced by a high-fat diet in C57BL/6J mice. Diabetologia 2011, 54, 922–934. [Google Scholar] [CrossRef] [Green Version]
- Perri, A.; Lofaro, D.; LA Russa, A.; Lupinacci, S.; Toteda, G.; Curti, A.; Urso, A.; Bonofiglio, R.; LA Russa, D.; Pellegrino, D.; et al. Proinflammatory profile of visceral adipose tissue and oxidative stress in severe obese patients carrying the variant rs4612666 C of NLRP3 gene. Minerva Endocrinol. 2021, 46, 309–316. [Google Scholar] [CrossRef]
- Wang, T.; Si, Y.; Shirihai, O.S.; Si, H.; Schultz, V.; Corkey, R.F.; Hu, L.; Deeney, J.T.; Guo, W.; Corkey, B.E. Respiration in adipocytes is inhibited by reactive oxygen species. Obesity 2010, 18, 1493–1502. [Google Scholar] [CrossRef]
- Gao, C.L.; Zhu, C.; Zhao, Y.P.; Chen, X.H.; Ji, C.B.; Zhang, C.M.; Zhu, J.G.; Xia, Z.K.; Tong, M.L.; Guo, X.R. Mitochondrial dysfunction is induced by high levels of glucose and free fatty acids in 3T3-L1 adipocytes. Mol. Cell Endocrinol. 2010, 320, 25–33. [Google Scholar] [CrossRef]
- Koh, E.H.; Park, J.Y.; Park, H.S.; Jeon, M.J.; Ryu, J.W.; Kim, M.; Kim, S.Y.; Kim, M.S.; Kim, S.W.; Park, I.S.; et al. Essential role of mitochondrial function in adiponectin synthesis in adipocytes. Diabetes 2007, 56, 2973–2981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marín-Royo, G.; Rodríguez, C.; Le Pape, A.; Jurado-López, R.; Luaces, M.; Antequera, A.; Martínez-González, J.; Souza-Neto, F.V.; Nieto, M.L.; Martínez-Martínez, E.; et al. The role of mitochondrial oxidative stress in the metabolic alterations in diet-induced obesity in rats. FASEB J. 2019, 33, 12060–12072. [Google Scholar] [CrossRef] [Green Version]
- Bogacka, I.; Xie, H.; Bray, G.A.; Smith, S.R. Pioglitazone induces mitochondrial biogenesis in human subcutaneous adipose tissue in vivo. Diabetes 2005, 54, 1392–1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heinonen, S.; Buzkova, J.; Muniandy, M.; Kaksonen, R.; Ollikainen, M.; Ismail, K.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Vuolteenaho, K.; et al. Impaired Mitochondrial Biogenesis in Adipose Tissue in Acquired Obesity. Diabetes 2015, 64, 3135–3145. [Google Scholar] [CrossRef] [Green Version]
- Powell-Wiley, T.M.; Poirier, P.; Burke, L.E.; Després, J.P.; Gordon-Larsen, P.; Lavie, C.J.; Lear, S.A.; Ndumele, C.E.; Neeland, I.J.; Sanders, P.; et al. Obesity and Cardiovascular Disease: A Scientific Statement from the American Heart Association. Circulation 2021, 143, e984–e1010. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Hu, X.; Hu, G.; Xu, C.; Jiang, H. Curcumin Attenuates Hydrogen Peroxide-Induced Premature Senescence via the Activation of SIRT1 in Human Umbilical Vein Endothelial Cells. Biol. Pharm. Bull. 2015, 38, 1134–1141. [Google Scholar] [CrossRef] [Green Version]
- Fleenor, B.S.; Sindler, A.L.; Marvi, N.K.; Howell, K.L.; Zigler, M.L.; Yoshizawa, M.; Seals, D.R. Curcumin ameliorates arterial dysfunction and oxidative stress with aging. Exp. Gerontol. 2013, 48, 269–276. [Google Scholar] [CrossRef] [Green Version]
- Sugawara, J.; Akazawa, N.; Miyaki, A.; Choi, Y.; Tanabe, Y.; Imai, T.; Maeda, S. Effect of endurance exercise training and curcumin intake on central arterial hemodynamics in postmenopausal women: Pilot study. Am. J. Hypertens. 2012, 25, 651–656. [Google Scholar] [CrossRef]
- Akazawa, N.; Choi, Y.; Miyaki, A.; Tanabe, Y.; Sugawara, J.; Ajisaka, R.; Maeda, S. Curcumin ingestion and exercise training improve vascular endothelial function in postmenopausal women. Nutr. Res. 2012, 32, 795–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos-Parker, J.R.; Strahler, T.R.; Bassett, C.J.; Bispham, N.Z.; Chonchol, M.B.; Seals, D.R. Curcumin supplementation improves vascular endothelial function in healthy middle-aged and older adults by increasing nitric oxide bioavailability and reducing oxidative stress. Aging 2017, 9, 187–208. [Google Scholar] [CrossRef] [Green Version]
- El-Far, A.H.; Elewa, Y.H.A.; Abdelfattah, E.A.; Alsenosy, A.A.; Atta, M.S.; Abou-Zeid, K.M.; Al Jaouni, S.K.; Mousa, S.A.; Noreldin, A.E. Thymoquinone and Curcumin Defeat Aging-Associated Oxidative Alterations Induced by D-Galactose in Rats’ Brain and Heart. Int. J. Mol. Sci. 2021, 22, 6839. [Google Scholar] [CrossRef] [PubMed]
- Ashrafizadeh, M.; Ahmadi, Z.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Curcumin Activates the Nrf2 Pathway and Induces Cellular Protection Against Oxidative Injury. Curr. Mol. Med. 2020, 20, 116–133. [Google Scholar] [CrossRef]
- González-Reyes, S.; Guzmán-Beltrán, S.; Medina-Campos, O.N.; Pedraza-Chaverri, J. Curcumin pretreatment induces Nrf2 and an antioxidant response and prevents hemin-induced toxicity in primary cultures of cerebellar granule neurons of rats. Oxid. Med. Cell. Longev. 2013, 2013, 801418. [Google Scholar] [CrossRef] [PubMed]
- Ren, L.; Zhan, P.; Wang, Q.; Wang, C.; Liu, Y.; Yu, Z.; Zhang, S. Curcumin upregulates the Nrf2 system by repressing inflammatory signaling-mediated Keap1 expression in insulin-resistant conditions. Biochem. Biophys. Res. Commun. 2019, 514, 691–698. [Google Scholar] [CrossRef] [PubMed]
- Ejaz, A.; Wu, D.; Kwan, P.; Meydani, M. Curcumin inhibits adipogenesis in 3T3-L1 adipocytes and angiogenesis and obesity in C57/BL mice. J. Nutr. 2009, 139, 919–925. [Google Scholar] [CrossRef]
- Wu, L.Y.; Chen, C.W.; Chen, L.K.; Chou, H.Y.; Chang, C.L.; Juan, C.C. Curcumin Attenuates Adipogenesis by Inducing Preadipocyte Apoptosis and Inhibiting Adipocyte Differentiation. Nutrients 2019, 11, 2307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weisberg, S.P.; Leibel, R.; Tortoriello, D.V. Dietary curcumin significantly improves obesity-associated inflammation and diabetes in mouse models of diabesity. Endocrinology 2008, 149, 3549–3558. [Google Scholar] [CrossRef] [Green Version]
- Shao, W.; Yu, Z.; Chiang, Y.; Yang, Y.; Chai, T.; Foltz, W.; Lu, H.; Fantus, I.G.; Jin, T. Curcumin prevents high fat diet induced insulin resistance and obesity via attenuating lipogenesis in liver and inflammatory pathway in adipocytes. PLoS ONE 2012, 7, e28784. [Google Scholar] [CrossRef]
- Song, Z.; Revelo, X.; Shao, W.; Tian, L.; Zeng, K.; Lei, H.; Sun, H.S.; Woo, M.; Winer, D.; Jin, T. Dietary Curcumin Intervention Targets Mouse White Adipose Tissue Inflammation and Brown Adipose Tissue UCP1 Expression. Obesity 2018, 26, 547–558. [Google Scholar] [CrossRef]
- Islam, T.; Koboziev, I.; Albracht-Schulte, K.; Mistretta, B.; Scoggin, S.; Yosofvand, M.; Moussa, H.; Zabet-Moghaddam, M.; Ramalingam, L.; Gunaratne, P.H.; et al. Curcumin Reduces Adipose Tissue Inflammation and Alters Gut Microbiota in Diet-Induced Obese Male Mice. Mol. Nutr. Food Res. 2021, 65, e2100274. [Google Scholar] [CrossRef]
- Kobori, M.; Takahashi, Y.; Takeda, H.; Takahashi, M.; Izumi, Y.; Akimoto, Y.; Sakurai, M.; Oike, H.; Nakagawa, T.; Itoh, M.; et al. Dietary Intake of Curcumin Improves eIF2 Signaling and Reduces Lipid Levels in the White Adipose Tissue of Obese Mice. Sci. Rep. 2018, 8, 9081. [Google Scholar] [CrossRef] [Green Version]
- Lone, J.; Choi, J.H.; Kim, S.W.; Yun, J.W. Curcumin induces brown fat-like phenotype in 3T3-L1 and primary white adipocytes. J. Nutr. Biochem. 2016, 27, 193–202. [Google Scholar] [CrossRef] [PubMed]
- Zhao, D.; Pan, Y.; Yu, N.; Bai, Y.; Ma, R.; Mo, F.; Zuo, J.; Chen, B.; Jia, Q.; Zhang, D.; et al. Curcumin improves adipocytes browning and mitochondrial function in 3T3-L1 cells and obese rodent model. R. Soc. Open Sci. 2021, 8, 200974. [Google Scholar] [CrossRef]
- Wang, S.; Wang, X.; Ye, Z.; Xu, C.; Zhang, M.; Ruan, B.; Wei, M.; Jiang, Y.; Zhang, Y.; Wang, L.; et al. Curcumin promotes browning of white adipose tissue in a norepinephrine-dependent way. Biochem. Biophys. Res. Commun. 2015, 466, 247–253. [Google Scholar] [CrossRef] [PubMed]
- Nishikawa, S.; Kamiya, M.; Aoyama, H.; Nomura, M.; Hyodo, T.; Ozeki, A.; Lee, H.; Takahashi, T.; Imaizumi, A.; Tsuda, T. Highly Dispersible and Bioavailable Curcumin but not Native Curcumin Induces Brown-Like Adipocyte Formation in Mice. Mol. Nutr. Food Res. 2018, 62, 1700731. [Google Scholar] [CrossRef]
- Scarpulla, R.C.; Vega, R.B.; Kelly, D.P. Transcriptional integration of mitochondrial biogenesis. Trends Endocrinol. Metab. 2012, 23, 459–466. [Google Scholar] [CrossRef] [Green Version]
- Di Pierro, F.; Bressan, A.; Ranaldi, D.; Rapacioli, G.; Giacomelli, L.; Bertuccioli, A. Potential role of bioavailable curcumin in weight loss and omental adipose tissue decrease: Preliminary data of a randomized, controlled trial in overweight people with metabolic syndrome. Preliminary study. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 4195–4202. [Google Scholar] [PubMed]
- Mohammadi, A.; Sahebkar, A.; Iranshahi, M.; Amini, M.; Khojasteh, R.; Ghayour-Mobarhan, M.; Ferns, G.A. Effects of supplementation with curcuminoids on dyslipidemia in obese patients: A randomized crossover trial. Phytother. Res. 2013, 27, 374–379. [Google Scholar] [CrossRef]
- Yang, Y.S.; Su, Y.F.; Yang, H.W.; Lee, Y.H.; Chou, J.I.; Ueng, K.C. Lipid-lowering effects of curcumin in patients with metabolic syndrome: A randomized, double-blind, placebo-controlled trial. Phytother. Res. 2014, 28, 1770–1777. [Google Scholar] [CrossRef] [PubMed]
- Alidadi, M.; Sahebkar, A.; Eslami, S.; Vakilian, F.; Jarahi, L.; Alinezhad-Namaghi, M.; Arabi, S.M.; Vakili, S.; Tohidinezhad, F.; Nikooiyan, Y.; et al. The Effect of Curcumin Supplementation on Pulse Wave Velocity in Patients with Metabolic Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Adv. Exp. Med. Biol. 2021, 1308, 1–11. [Google Scholar] [PubMed]
- Chuengsamarn, S.; Rattanamongkolgul, S.; Phonrat, B.; Tungtrongchitr, R.; Jirawatnotai, S. Reduction of atherogenic risk in patients with type 2 diabetes by curcuminoid extract: A randomized controlled trial. J. Nutr. Biochem. 2014, 25, 144–150. [Google Scholar] [CrossRef]
- Akbari, M.; Lankarani, K.B.; Tabrizi, R.; Ghayour-Mobarhan, M.; Peymani, P.; Ferns, G.; Ghaderi, A.; Asemi, Z. The Effects of Curcumin on Weight Loss Among Patients With Metabolic Syndrome and Related Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Pharmacol. 2019, 10, 649. [Google Scholar] [CrossRef] [Green Version]
- Quiles, J.L.; Mesa, M.D.; Ramírez-Tortosa, C.L.; Aguilera, C.M.; Battino, M.; Gil, A.; Ramírez-Tortosa, M.C. Curcuma longa extract supplementation reduces oxidative stress and attenuates aortic fatty streak development in rabbits. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 1225–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olszanecki, R.; Jawień, J.; Gajda, M.; Mateuszuk, L.; Gebska, A.; Korabiowska, M.; Chłopicki, S.; Korbut, R. Effect of curcumin on atherosclerosis in apoE/LDLR-double knockout mice. J. Physiol. Pharmacol. 2005, 56, 627–635. [Google Scholar]
- Shin, S.K.; Ha, T.Y.; McGregor, R.A.; Choi, M.S. Long-term curcumin administration protects against atherosclerosis via hepatic regulation of lipoprotein cholesterol metabolism. Mol. Nutr. Food Res. 2011, 55, 1829–1840. [Google Scholar] [CrossRef]
- Coban, D.; Milenkovic, D.; Chanet, A.; Khallou-Laschet, J.; Sabbe, L.; Palagani, A.; Vanden Berghe, W.; Mazur, A.; Morand, C. Dietary curcumin inhibits atherosclerosis by affecting the expression of genes involved in leukocyte adhesion and transendothelial migration. Mol. Nutr. Food Res. 2012, 56, 1270–1281. [Google Scholar] [CrossRef]
- Hasan, S.T.; Zingg, J.M.; Kwan, P.; Noble, T.; Smith, D.; Meydani, M. Curcumin modulation of high fat diet-induced atherosclerosis and steatohepatosis in LDL receptor deficient mice. Atherosclerosis 2014, 232, 40–51. [Google Scholar] [CrossRef] [PubMed]
- Wan, Q.; Liu, Z.Y.; Yang, Y.P.; Liu, S.M. Effect of curcumin on inhibiting atherogenesis by down-regulating lipocalin-2 expression in apolipoprotein E knockout mice. Biomed. Mater. Eng. 2016, 27, 577–587. [Google Scholar] [CrossRef]
- Zou, J.; Zhang, S.; Li, P.; Zheng, X.; Feng, D. Supplementation with curcumin inhibits intestinal cholesterol absorption and prevents atherosclerosis in high-fat diet-fed apolipoprotein E knockout mice. Nutr. Res. 2018, 56, 32–40. [Google Scholar] [CrossRef]
- Ramírez-Tortosa, M.C.; Mesa, M.D.; Aguilera, M.C.; Quiles, J.L.; Baró, L.; Ramirez-Tortosa, C.L.; Martinez-Victoria, E.; Gil, A. Oral administration of a turmeric extract inhibits LDL oxidation and has hypocholesterolemic effects in rabbits with experimental atherosclerosis. Atherosclerosis 1999, 147, 371–378. [Google Scholar] [CrossRef]
- Li, X.; Lu, Y.; Sun, Y.; Zhang, Q. Effect of curcumin on permeability of coronary artery and expression of related proteins in rat coronary atherosclerosis heart disease model. Int. J. Clin. Exp. Pathol. 2015, 8, 7247–7253. [Google Scholar]
- Quiles, J.L.; Aguilera, C.; Mesa, M.D.; Ramírez-Tortosa, M.C.; Baró, L.; Gil, A. An ethanolic-aqueous extract of Curcuma longa decreases the susceptibility of liver microsomes and mitochondria to lipid peroxidation in atherosclerotic rabbits. Biofactors 1998, 8, 51–57. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Little, P.J.; Xu, S.; Kamato, D. Curcumin Inhibits Lysophosphatidic Acid Mediated MCP-1 Expression via Blocking ROCK Signalling. Molecules 2021, 26, 2320. [Google Scholar] [CrossRef]
- Nirmala, C.; Puvanakrishnan, R. Protective role of curcumin against isoproterenol induced myocardial infarction in rats. Mol. Cell. Biochem. 1996, 159, 85–93. [Google Scholar] [CrossRef] [PubMed]
- Rahnavard, M.; Hassanpour, M.; Ahmadi, M.; Heidarzadeh, M.; Amini, H.; Javanmard, M.Z.; Nouri, M.; Rahbarghazi, R.; Safaie, N. Curcumin ameliorated myocardial infarction by inhibition of cardiotoxicity in the rat model. J. Cell. Biochem. 2019, 120, 11965–11972. [Google Scholar] [CrossRef]
- Duan, W.; Yang, Y.; Yan, J.; Yu, S.; Liu, J.; Zhou, J.; Zhang, J.; Jin, Z.; Yi, D. The effects of curcumin post-treatment against myocardial ischemia and reperfusion by activation of the JAK2/STAT3 signaling pathway. Basic Res. Cardiol. 2012, 107, 263. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Duan, W.; Lin, Y.; Yi, W.; Liang, Z.; Yan, J.; Wang, N.; Deng, C.; Zhang, S.; Li, Y.; et al. SIRT1 activation by curcumin pretreatment attenuates mitochondrial oxidative damage induced by myocardial ischemia reperfusion injury. Free Radic. Biol. Med. 2013, 65, 667–679. [Google Scholar] [CrossRef]
- Liu, K.; Chen, H.; You, Q.S.; Ye, Q.; Wang, F.; Wang, S.; Zhang, S.L.; Yu, K.J.; Lu, Q. Curcumin attenuates myocardial ischemia-reperfusion injury. Oncotarget 2017, 8, 112051–112059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, N.P.; Wang, Z.F.; Tootle, S.; Philip, T.; Zhao, Z.Q. Curcumin promotes cardiac repair and ameliorates cardiac dysfunction following myocardial infarction. Br. J. Pharmacol. 2012, 167, 1550–1562. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Wu, M.; Tang, L.; Pan, Y.; Liu, Z.; Zeng, C.; Wang, J.; Wei, T.; Liang, G. Novel curcumin analogue 14p protects against myocardial ischemia reperfusion injury through Nrf2-activating anti-oxidative activity. Toxicol. Appl. Pharmacol. 2015, 282, 175–183. [Google Scholar] [CrossRef] [PubMed]
- Liao, C.L.; Liu, Y.; Huang, M.Z.; Liu, H.Y.; Ye, Z.L.; Su, Q. Myocardial ischemia reperfusion injury is alleviated by curcumin-peptide hydrogel via upregulating autophagy and protecting mitochondrial function. Stem. Cell Res. Ther. 2021, 12, 89. [Google Scholar] [CrossRef]
- Wu, H.J.; Zhang, K.; Ma, J.J.; Wang, L.; Zhuang, Y. Mechanism of curcumin against myocardial ischaemia-reperfusion injury based on the P13K/Akt/mTOR signalling pathway. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 5490–5499. [Google Scholar]
- Boarescu, P.M.; Boarescu, I.; Bocșan, I.C.; Pop, R.M.; Gheban, D.; Bulboacă, A.E.; Nicula, C.; Râjnoveanu, R.M.; Bolboacă, S.D. Curcumin Nanoparticles Protect against Isoproterenol Induced Myocardial Infarction by Alleviating Myocardial Tissue Oxidative Stress, Electrocardiogram, and Biological Changes. Molecules 2019, 24, 2802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, H.; Wang, C.; Qiao, Z.; Xu, Y. Protective effect of curcumin against myocardium injury in ischemia reperfusion rats. Pharm. Biol. 2017, 55, 1144–1148. [Google Scholar] [CrossRef] [PubMed]
- Lv, F.H.; Yin, H.L.; He, Y.Q.; Wu, H.M.; Kong, J.; Chai, X.Y.; Zhang, S.R. Effects of curcumin on the apoptosis of cardiomyocytes and the expression of NF-κB, PPAR-γ and Bcl-2 in rats with myocardial infarction injury. Exp. Ther. Med. 2016, 12, 3877–3884. [Google Scholar] [CrossRef] [Green Version]
- Yan, S.; Zhou, M.; Zheng, X.; Xing, Y.; Dong, J.; Yan, M.; Li, R. Anti-Inflammatory Effect of Curcumin on the Mouse Model of Myocardial Infarction through Regulating Macrophage Polarization. Mediat. Inflamm. 2021, 2021, 9976912. [Google Scholar] [CrossRef]
- González-Salazar, A.; Molina-Jijón, E.; Correa, F.; Zarco-Márquez, G.; Calderón-Oliver, M.; Tapia, E.; Zazueta, C.; Pedraza-Chaverri, J. Curcumin protects from cardiac reperfusion damage by attenuation of oxidant stress and mitochondrial dysfunction. Cardiovasc. Toxicol. 2011, 11, 357–364. [Google Scholar] [CrossRef]
- Tang, B.L. Sirt1 and the Mitochondria. Mol. Cells 2016, 39, 87–95. [Google Scholar]
- Packer, M. Cardioprotective Effects of Sirtuin-1 and Its Downstream Effectors: Potential Role in Mediating the Heart Failure Benefits of SGLT2 (Sodium-Glucose Cotransporter 2) Inhibitors. Circ. Heart Fail. 2020, 13, e007197. [Google Scholar] [CrossRef]
- Bravo-San Pedro, J.M.; Kroemer, G.; Galluzzi, L. Autophagy and Mitophagy in Cardiovascular Disease. Circ. Res. 2017, 120, 1812–1824. [Google Scholar] [CrossRef]
- Yu, W.; Qin, J.; Chen, C.; Fu, Y.; Wang, W. Moderate calorie restriction attenuates age-associated alterations and improves cardiac function by increasing SIRT1 and SIRT3 expression. Mol. Med. Rep. 2018, 18, 4087–4094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dolinsky, V.W.; Dyck, J.R. Calorie restriction and resveratrol in cardiovascular health and disease. Biochim. Biophys. Acta 2011, 1812, 1477–1489. [Google Scholar] [CrossRef] [Green Version]
- Chung, K.W.; Chung, H.Y. The Effects of Calorie Restriction on Autophagy: Role on Aging Intervention. Nutrients 2019, 11, 2923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blagosklonny, M.V. Calorie restriction: Decelerating mTOR-driven aging from cells to organisms (including humans). Cell Cycle 2010, 9, 683–688. [Google Scholar] [CrossRef]
- Wan, W.; You, Z.; Xu, Y.; Zhou, L.; Guan, Z.; Peng, C.; Wong, C.C.L.; Su, H.; Zhou, T.; Xia, H.; et al. mTORC1 Phosphorylates Acetyltransferase p300 to Regulate Autophagy and Lipogenesis. Mol. Cell. 2017, 68, 323–335.e326. [Google Scholar] [CrossRef] [Green Version]
- Yang, Z.; Ming, X.F. mTOR signalling: The molecular interface connecting metabolic stress, aging and cardiovascular diseases. Obes. Rev. 2012, 13 (Suppl. 2), 58–68. [Google Scholar] [CrossRef]
- Hofer, S.J.; Davinelli, S.; Bergmann, M.; Scapagnini, G.; Madeo, F. Caloric Restriction Mimetics in Nutrition and Clinical Trials. Front. Nutr. 2021, 8, 717343. [Google Scholar] [CrossRef]
- Voglhuber, J.; Ljubojevic-Holzer, S.; Abdellatif, M.; Sedej, S. Targeting Cardiovascular Risk Factors Through Dietary Adaptations and Caloric Restriction Mimetics. Front. Nutr. 2021, 8, 758058. [Google Scholar] [CrossRef] [PubMed]
- Balasubramanyam, K.; Varier, R.A.; Altaf, M.; Swaminathan, V.; Siddappa, N.B.; Ranga, U.; Kundu, T.K. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. J. Biol. Chem. 2004, 279, 51163–51171. [Google Scholar] [CrossRef] [Green Version]
- Han, J.; Pan, X.Y.; Xu, Y.; Xiao, Y.; An, Y.; Tie, L.; Pan, Y.; Li, X.J. Curcumin induces autophagy to protect vascular endothelial cell survival from oxidative stress damage. Autophagy 2012, 8, 812–825. [Google Scholar] [CrossRef] [Green Version]
- Guo, S.; Long, M.; Li, X.; Zhu, S.; Zhang, M.; Yang, Z. Curcumin activates autophagy and attenuates oxidative damage in EA.hy926 cells via the Akt/mTOR pathway. Mol. Med. Rep. 2016, 13, 2187–2193. [Google Scholar] [CrossRef] [PubMed]
- Ye, M.; Qiu, H.; Cao, Y.; Zhang, M.; Mi, Y.; Yu, J.; Wang, C. Curcumin Improves Palmitate-Induced Insulin Resistance in Human Umbilical Vein Endothelial Cells by Maintaining Proteostasis in Endoplasmic Reticulum. Front. Pharmacol. 2017, 8, 148. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.; Zhu, Y.; Li, K.; Liao, B.; Wang, F.; Shao, L.; Huang, L.; Bai, D. Curcumin-mediated Photodynamic Therapy Inhibits the Phenotypic Transformation, Migration, and Foaming of Oxidized Low-density Lipoprotein-treated Vascular Smooth Muscle Cells by Promoting Autophagy. J. Cardiovasc. Pharmacol. 2021, 78, 308–318. [Google Scholar] [CrossRef]
- Yang, K.; Xu, C.; Li, X.; Jiang, H. Combination of D942 with curcumin protects cardiomyocytes from ischemic damage through promoting autophagy. J. Cardiovasc. Pharmacol. Ther. 2013, 18, 570–581. [Google Scholar] [CrossRef]
- Galluzzi, L.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cecconi, F.; Choi, A.M.; Chu, C.T.; Codogno, P.; Colombo, M.I.; et al. Molecular definitions of autophagy and related processes. EMBO J. 2017, 36, 1811–1836. [Google Scholar] [CrossRef]
- Wang, W.; Xu, J. Curcumin Attenuates Cerebral Ischemia-reperfusion Injury Through Regulating Mitophagy and Preserving Mitochondrial Function. Curr. Neurovasc. Res. 2020, 17, 113–122. [Google Scholar] [CrossRef]
- Oshima, Y.; Fujio, Y.; Nakanishi, T.; Itoh, N.; Yamamoto, Y.; Negoro, S.; Tanaka, K.; Kishimoto, T.; Kawase, I.; Azuma, J. STAT3 mediates cardioprotection against ischemia/reperfusion injury through metallothionein induction in the heart. Cardiovasc. Res. 2005, 65, 428–435. [Google Scholar] [CrossRef] [PubMed]
- Heusch, G. 25 years of remote ischemic conditioning: From laboratory curiosity to clinical outcome. Basic Res. Cardiol. 2018, 113, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kleinbongard, P.; Gedik, N.; Kirca, M.; Stoian, L.; Frey, U.; Zandi, A.; Thielmann, M.; Jakob, H.; Peters, J.; Kamler, M.; et al. Mitochondrial and Contractile Function of Human Right Atrial Tissue in Response to Remote Ischemic Conditioning. J. Am. Heart Assoc. 2018, 7, e009540. [Google Scholar] [CrossRef] [Green Version]
- Heusch, G.; Musiolik, J.; Gedik, N.; Skyschally, A. Mitochondrial STAT3 activation and cardioprotection by ischemic postconditioning in pigs with regional myocardial ischemia/reperfusion. Circ. Res. 2011, 109, 1302–1308. [Google Scholar] [CrossRef] [Green Version]
- Boengler, K.; Buechert, A.; Heinen, Y.; Roeskes, C.; Hilfiker-Kleiner, D.; Heusch, G.; Schulz, R. Cardioprotection by ischemic postconditioning is lost in aged and STAT3-deficient mice. Circ. Res. 2008, 102, 131–135. [Google Scholar] [CrossRef] [Green Version]
- Kumar, A.; Harsha, C.; Parama, D.; Girisa, S.; Daimary, U.D.; Mao, X.; Kunnumakkara, A.B. Current clinical developments in curcumin-based therapeutics for cancer and chronic diseases. Phytother. Res. 2021, 35, 6768–6801. [Google Scholar] [CrossRef] [PubMed]
- Vareed, S.K.; Kakarala, M.; Ruffin, M.T.; Crowell, J.A.; Normolle, D.P.; Djuric, Z.; Brenner, D.E. Pharmacokinetics of curcumin conjugate metabolites in healthy human subjects. Cancer Epidemiol. Biomark. Prev. 2008, 17, 1411–1417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srinivas, P.S. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med. 1998, 64, 353–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, P.; Li, H.; Lin, Z.; Luo, H.; Luo, W. Comparing the Effect of Piperine and Ilepcimide on the Pharmacokinetics of Curcumin in SD Rats. Front. Pharmacol. 2021, 12, 725362. [Google Scholar] [CrossRef]
- Grilc, N.K.; Sova, M.; Kristl, J. Drug Delivery Strategies for Curcumin and Other Natural Nrf2 Modulators of Oxidative Stress-Related Diseases. Pharmaceutics 2021, 13, 2137. [Google Scholar] [CrossRef]
- Sohn, S.I.; Priya, A.; Balasubramaniam, B.; Muthuramalingam, P.; Sivasankar, C.; Selvaraj, A.; Valliammai, A.; Jothi, R.; Pandian, S. Biomedical Applications and Bioavailability of Curcumin-An Updated Overview. Pharmaceutics 2021, 13, 2102. [Google Scholar] [CrossRef]
- Jamwal, R. Bioavailable curcumin formulations: A review of pharmacokinetic studies in healthy volunteers. J. Integr. Med. 2018, 16, 367–374. [Google Scholar] [CrossRef]
- Freedman, N.D.; Park, Y.; Abnet, C.C.; Hollenbeck, A.R.; Sinha, R. Association of coffee drinking with total and cause-specific mortality. N. Engl. J. Med. 2012, 366, 1891–1904. [Google Scholar] [CrossRef] [PubMed]
- Gunter, M.J.; Murphy, N.; Cross, A.J.; Dossus, L.; Dartois, L.; Fagherazzi, G.; Kaaks, R.; Kühn, T.; Boeing, H.; Aleksandrova, K.; et al. Coffee Drinking and Mortality in 10 European Countries: A Multinational Cohort Study. Ann. Intern. Med. 2017, 167, 236–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stevens, L.M.; Linstead, E.; Hall, J.L.; Kao, D.P. Association Between Coffee Intake and Incident Heart Failure Risk: A Machine Learning Analysis of the FHS, the ARIC Study, and the CHS. Circ. Heart Fail. 2021, 14, e006799. [Google Scholar] [CrossRef] [PubMed]
- Heiss, C.; Spyridopoulos, I.; Haendeler, J. Interventions to slow cardiovascular aging: Dietary restriction, drugs and novel molecules. Exp. Gerontol. 2018, 109, 108–118. [Google Scholar] [CrossRef]
- Parmenter, B.H.; Croft, K.D.; Hodgson, J.M.; Dalgaard, F.; Bondonno, C.P.; Lewis, J.R.; Cassidy, A.; Scalbert, A.; Bondonno, N.P. An overview and update on the epidemiology of flavonoid intake and cardiovascular disease risk. Food Funct. 2020, 11, 6777–6806. [Google Scholar] [CrossRef]
- Harley, C.B.; Liu, W.; Flom, P.L.; Raffaele, J.M. A natural product telomerase activator as part of a health maintenance program: Metabolic and cardiovascular response. Rejuvenation Res. 2013, 16, 386–395. [Google Scholar] [CrossRef] [PubMed]
Model | Curcumin Dose and Application Route | Curcumin Effects | Ref. |
---|---|---|---|
New Zealand rabbits on HFD | 1.66 and 3.2 mg/kg bw turmeric hydroalcoholic extract (10% curcumin) oral for 7 weeks | intracellular membrane lipid peroxidation ↓ | [147] |
New Zealand rabbits on HFD | 1.66 and 3.2 mg/kg bw turmeric hydroalcoholic extract (10% curcumin) oral for 7 weeks | total cholesterol ↓ LDL-cholesterol ↓ LDL-triglycerides ↓ LDL-phospholipids ↓ LDL lipid peroxidation ↓ | [145] |
New Zealand rabbits on HFD | 1.66 mg/kg bw turmeric hydroalcoholic extract (10% curcumin) oral for 10, 20, and 30 days | lesion size ↓ plasma lipid peroxidation ↓ | [138] |
Wistar rats on HFD plus intraperitoneal vitamin D3 injection | 100 mg/kg bw curcumin per day via oral gavage for 4 weeks | triglycerides ↓ total cholesterol ↓ LDL-cholesterol ↓ HDL-cholesterol ↑ arterial permeability ↓ MMP9 ↓ TNFα, CRP ↓ | [146] |
C57BL/6J x 129/SvJ Apoe−/− Ldlr−/− mice on HFD | 0.3 mg/day curcumin in chow for 16 weeks | lesion size ↓ | [139] |
C57BL/6J Ldlr−/− mice on HFD | 500, 1000, and 1500 mg/kg bw curcumin in chow for 16 weeks | lesion size ↓ IL-6, MCP1 ↓ CD36 ↓ | [142] |
C57BL/6J Ldlr−/− mice on HFD | 0.02% (w/w) curcumin in chow for 18 weeks | lesion size ↓ triglycerides ↓ total cholesterol ↓ LDL-cholesterol ↓ HDL-cholesterol ↑ HMG-CoA reductase ↓ CRP ↓ | [140] |
C57BL/6J Apoe−/− mice on HFD | 0.1% (w/w) curcumin in chow for 16 weeks | lesion size ↓ total cholesterol ↓ LDL-cholesterol ↓ | [144] |
C57BL/6J Apoe−/− mice on HFD | 0.2% (w/w) curcumin in chow for 16 weeks | lesion size ↓ macrophage infiltration ↓ IκB ↑ | [141] |
C57BL/6J Apoe−/− mice on HFD | 40, 60 and 80 mg/kg bw curcumin per day via oral gavage for 12 weeks | lesion size ↓ triglycerides ↓ total cholesterol ↓ LDL-cholesterol ↓ TNFα, CRP, IL-6, LCN2 ↓ | [143] |
Model | Curcumin Dose and Application Route | Curcumin Effects | Ref. |
---|---|---|---|
Wistar rats subcutaneous isoproterenol injection | 25, 50, 100, 200 mg/kg curcumin per day via oral gavage starting 2 days before isoproterenol | heart function ↑ CK, LDH ↓ anti-oxidative enzymes ↑ lipid peroxidation ↓ | [149] |
Wistar rats subcutaneous isoproterenol injection | 50 mg/kg bw curcumin per day via oral gavage for 9 days starting with isoproterenol | scar size ↓ CK, LDH ↓ apoptosis ↓ SOD ↑ oxidative stress ↓ | [150] |
Wistar-Bratislava rats subcutaneous isoproterenol injection | 100, 150 and 200 mg/kg bw curcumin per day via oral gavage for 15 days starting with isoproterenol | heart function ↑ CK, LDH ↓ oxidative stress ↓ | [158] |
Sprague–Dawley rats LAD ligation | 200 mg/kg bw curcumin per day via oral gavage starting 10 days before MI | infarct size ↓ CK, LDH ↓ BCL2 ↑ BAX ↓ SIRT1 ↑ | [152] |
Sprague–Dawley rats LAD ligation | 10, 20 and 30 mg/kg bw curcumin per day via oral gavage starting 20 days before MI | infarct size ↓ heart function ↑ BCL2 ↑ BAX ↓ | [159] |
Sprague–Dawley rats LAD ligation | 25, 50 and 100 mg/kg bw curcumin single intraperitoneal bolus injection 30 min before MI | infarct size ↓ CK, LDH ↓ SOD, GSH ↑ BCL2 ↑ BAX ↓ oxidative stress ↓ | [157] |
Sprague-Dawley rats LAD ligation | 20 µL 40 μM curcumin or curcumin hydrogel injected into the ventricular wall during ischemia | infarct size ↓ heart function ↑ apoptosis ↓ anti-oxidative enzymes ↑ phospho-JAK2/STAT3 ↑ | [156] |
C57BL/6 mice LAD ligation | 100 mg/kg bw curcumin or 10 mg/kg bw curcumin analog 14p per day via oral gavage starting 7 days before MI | infarct size ↓ CK ↓ apoptosis ↓ BCL2/BAX ratio ↑ oxidative stress ↓ NRF2 ↑ | [155] |
Sprague-Dawley rats LAD ligation | 150 mg/kg bw curcumin per day in peanut paste for 7, 21, or 42 days starting after ischemia | infarct size ↓ heart function ↑ scar size ↓ MMP2, MMP9 ↓ Collagen I, III, IV ↓ myofibroblasts ↓ | [154] |
Sprague-Dawley rats LAD ligation | 150 mg/kg bw curcumin per day in peanut paste for 28 days starting after ischemia | apoptosis ↓ BCL2 ↑ NFκB p65 ↓ | [160] |
C57BL/6 mice LAD ligation | 100 mg/kg/day curcumin injected intraperitoneally for 6 weeks starting after ischemia | heart function ↑ fibrosis ↓ Collagen I ↓ TNFα, IL-6, IL-1β ↓ M1 macrophages ↓ M2 macrophages ↑ | [161] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cox, F.F.; Misiou, A.; Vierkant, A.; Ale-Agha, N.; Grandoch, M.; Haendeler, J.; Altschmied, J. Protective Effects of Curcumin in Cardiovascular Diseases—Impact on Oxidative Stress and Mitochondria. Cells 2022, 11, 342. https://doi.org/10.3390/cells11030342
Cox FF, Misiou A, Vierkant A, Ale-Agha N, Grandoch M, Haendeler J, Altschmied J. Protective Effects of Curcumin in Cardiovascular Diseases—Impact on Oxidative Stress and Mitochondria. Cells. 2022; 11(3):342. https://doi.org/10.3390/cells11030342
Chicago/Turabian StyleCox, Fiona Frederike, Angelina Misiou, Annika Vierkant, Niloofar Ale-Agha, Maria Grandoch, Judith Haendeler, and Joachim Altschmied. 2022. "Protective Effects of Curcumin in Cardiovascular Diseases—Impact on Oxidative Stress and Mitochondria" Cells 11, no. 3: 342. https://doi.org/10.3390/cells11030342
APA StyleCox, F. F., Misiou, A., Vierkant, A., Ale-Agha, N., Grandoch, M., Haendeler, J., & Altschmied, J. (2022). Protective Effects of Curcumin in Cardiovascular Diseases—Impact on Oxidative Stress and Mitochondria. Cells, 11(3), 342. https://doi.org/10.3390/cells11030342