51
|
Zhao Q, Zhou J, Li F, Guo S, Zhang L, Li J, Qi Q, Shi Y. The Role and Therapeutic Perspectives of Sirtuin 3 in Cancer Metabolism Reprogramming, Metastasis, and Chemoresistance. Front Oncol 2022; 12:910963. [PMID: 35832551 PMCID: PMC9272524 DOI: 10.3389/fonc.2022.910963] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022] Open
Abstract
Sirtuin 3 (SIRT3), the nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, acts as a metabolic modulator mainly located in mitochondria via regulating the process of the relevant biochemical processes by targeting crucial mediators. Recently, owing to its dual role in cancer, SIRT3 has attracted extensive attention. Cancer cells have different metabolic patterns from normal cells, and SIRT3-mediated metabolism reprogramming could be critical in the cancer context, which is closely related to the mechanism of metabolism reprogramming, metastasis, and chemoresistance in tumor cells. Therefore, it is crucial to elucidate the relevant pathological mechanisms and take appropriate countermeasures for the progression of clinical strategies to inhibit the development of cancer. In this review, existing available data on the regulation of cancer metabolism reprogramming, metastasis, and chemoresistance progression of SIRT3 are detailed, as well as the status quo of SIRT3 small molecule modulators is updated in the application of cancer therapy, aiming to highlight strategies directly targeting SIRT3-mediated tumor-suppressing and tumor-promoting, and provide new approaches for therapy application. Furthermore, we offer an effective evidence-based basis for the evolvement of potential personalized therapy management strategies for SIRT3 in cancer settings.
Collapse
Affiliation(s)
- QingYi Zhao
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhou
- Department of Acupuncture and Moxibustion, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Li
- Department of Acupuncture and Moxibustion, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sen Guo
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhang
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Li
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin Qi
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Outpatient Department, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- *Correspondence: Qin Qi, ; Yin Shi,
| | - Yin Shi
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Outpatient Department, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- *Correspondence: Qin Qi, ; Yin Shi,
| |
Collapse
|
52
|
Stojanovic D, Mitic V, Stojanovic M, Milenkovic J, Ignjatovic A, Milojkovic M. The Scientific Rationale for the Introduction of Renalase in the Concept of Cardiac Fibrosis. Front Cardiovasc Med 2022; 9:845878. [PMID: 35711341 PMCID: PMC9193824 DOI: 10.3389/fcvm.2022.845878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
Cardiac fibrosis represents a redundant accumulation of extracellular matrix proteins, resulting from a cascade of pathophysiological events involved in an ineffective healing response, that eventually leads to heart failure. The pathophysiology of cardiac fibrosis involves various cellular effectors (neutrophils, macrophages, cardiomyocytes, fibroblasts), up-regulation of profibrotic mediators (cytokines, chemokines, and growth factors), and processes where epithelial and endothelial cells undergo mesenchymal transition. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. The most effective anti-fibrotic strategy will have to incorporate the specific targeting of the diverse cells, pathways, and their cross-talk in the pathogenesis of cardiac fibroproliferation. Additionally, renalase, a novel protein secreted by the kidneys, is identified. Evidence demonstrates its cytoprotective properties, establishing it as a survival element in various organ injuries (heart, kidney, liver, intestines), and as a significant anti-fibrotic factor, owing to its, in vitro and in vivo demonstrated pleiotropy to alleviate inflammation, oxidative stress, apoptosis, necrosis, and fibrotic responses. Effective anti-fibrotic therapy may seek to exploit renalase’s compound effects such as: lessening of the inflammatory cell infiltrate (neutrophils and macrophages), and macrophage polarization (M1 to M2), a decrease in the proinflammatory cytokines/chemokines/reactive species/growth factor release (TNF-α, IL-6, MCP-1, MIP-2, ROS, TGF-β1), an increase in anti-apoptotic factors (Bcl2), and prevention of caspase activation, inflammasome silencing, sirtuins (1 and 3) activation, and mitochondrial protection, suppression of epithelial to mesenchymal transition, a decrease in the pro-fibrotic markers expression (’α-SMA, collagen I, and III, TIMP-1, and fibronectin), and interference with MAPKs signaling network, most likely as a coordinator of pro-fibrotic signals. This review provides the scientific rationale for renalase’s scrutiny regarding cardiac fibrosis, and there is great anticipation that these newly identified pathways are set to progress one step further. Although substantial progress has been made, indicating renalase’s therapeutic promise, more profound experimental work is required to resolve the accurate underlying mechanisms of renalase, concerning cardiac fibrosis, before any potential translation to clinical investigation.
Collapse
Affiliation(s)
- Dijana Stojanovic
- Institute of Pathophysiology, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Valentina Mitic
- Department of Cardiovascular Rehabilitation, Institute for Treatment and Rehabilitation "Niska Banja", Niska Banja, Serbia
| | - Miodrag Stojanovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Niš, Niš, Serbia.,Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, Niš, Serbia
| | - Jelena Milenkovic
- Institute of Pathophysiology, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Aleksandra Ignjatovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Niš, Niš, Serbia.,Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, Niš, Serbia
| | - Maja Milojkovic
- Institute of Pathophysiology, Faculty of Medicine, University of Niš, Niš, Serbia
| |
Collapse
|
53
|
Tomczyk MM, Cheung KG, Xiang B, Tamanna N, Fonseca Teixeira AL, Agarwal P, Kereliuk SM, Spicer V, Lin L, Treberg J, Tong Q, Dolinsky VW. Mitochondrial Sirtuin-3 (SIRT3) Prevents Doxorubicin-Induced Dilated Cardiomyopathy by Modulating Protein Acetylation and Oxidative Stress. Circ Heart Fail 2022; 15:e008547. [PMID: 35418250 PMCID: PMC9117478 DOI: 10.1161/circheartfailure.121.008547] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.
Collapse
Affiliation(s)
- Mateusz M Tomczyk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Kyle G Cheung
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Bo Xiang
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Nahid Tamanna
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Ana L Fonseca Teixeira
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Prasoon Agarwal
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,KTH Royal Institute of Technology, School of Electrical Engineering and Computer Science, Stockholm, Sweden (P.A.).,Science for Life Laboratory, Solna, Sweden (P.A.)
| | - Stephanie M Kereliuk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Victor Spicer
- Department of Internal Medicine (V.S.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,Manitoba Center for Proteomics and Systems Biology, Winnipeg, Canada (V.S.)
| | - Ligen Lin
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.).,Institute of Chinese Medical Sciences, University of Macau, China (L.L.)
| | - Jason Treberg
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.)
| | - Vernon W Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| |
Collapse
|
54
|
Huang A, Yang F, Cheng P, Liao D, Zhou L, Ji X, Peng D, Zhang L, Cheng T, Ma L, Xia X. Honokiol attenuate the arsenic trioxide-induced cardiotoxicity by reducing the myocardial apoptosis. Pharmacol Res Perspect 2022; 10:e00914. [PMID: 35171536 PMCID: PMC8848632 DOI: 10.1002/prp2.914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023] Open
Abstract
Despite advantages of arsenic trioxide (ATO) in oncological practice, its clinical applications have been hampered by severe cardiotoxicity. The general mechanism of ATO-induced cardiotoxicity has been attributed to its damage to mitochondria, resulting in cardiac remodeling. Honokiol (HKL) is a naturally occurring compound derived from Magnolia bark. Previous studies have demonstrated that HKL exerts cardio-protective effects on ischemia/reperfusion (I/R) or chemical-induced cardiotoxicity by counteracting the toxic effects on mitochondria. The present study was conducted to investigate whether HKL pretreatment protects against ATO-induced cardiac oxidative damage and cell death. For the in vitro study, we evaluated the effects of ATO and/or Honokiol on reactive oxygen species (ROS) production and apoptosis induction in primary cultured cardiomyocytes; for the in vivo study, BALB/c mice were administrated with ATO and/or HKL for a period of 4 weeks, myocardial apoptosis, cardiac function, and cardiac remodeling (cardiac hypertrophy and cardiac fibrosis) were assessed at the end of administration. Our results demonstrated Honokiol pretreatment alleviated the ATO-induced boost in ROS concentration and the following apoptosis induction in primary cultured cardiomyocytes. In the mouse model, Honokiol pretreatment ameliorated ATO-induced myocardial apoptosis, cardiac dysfunction, and cardiac remodeling. Collectively, these results indicated that Honokiol provide a protection against ATO-induced cardiotoxicity by reducing mitochondrial damage. In addition, given that Honokiol has shown considerable suppressive effects on leukemia cells, our data also imply that ATO and Honokiol combination may possibly be a superior avenue in leukemia therapy.
Collapse
Affiliation(s)
- An‐Liang Huang
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Fan Yang
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Ping Cheng
- State Key Lab of BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople’s Republic of China
| | - Dian‐ying Liao
- Department of PathologyWest China HospitalChengduSichuanPeople’s Republic of China
| | - Li Zhou
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Xing‐Li Ji
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Dou‐Dou Peng
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Li Zhang
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Ting‐Ting Cheng
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Li Ma
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| | - Xian‐Gen Xia
- Department of PathologyChengdu Fifth People’s HospitalChengduSichuanPeople’s Republic of China
- Department of PathologyThe Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese MedicineChengduSichuanPeople’s Republic of China
| |
Collapse
|
55
|
Murugasamy K, Munjal A, Sundaresan NR. Emerging Roles of SIRT3 in Cardiac Metabolism. Front Cardiovasc Med 2022; 9:850340. [PMID: 35369299 PMCID: PMC8971545 DOI: 10.3389/fcvm.2022.850340] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2022] [Indexed: 12/17/2022] Open
Abstract
The heart is a highly metabolically active organ that predominantly utilizes fatty acids as an energy substrate. The heart also derives some part of its energy by oxidation of other substrates, including glucose, lactose, amino acids and ketones. The critical feature of cardiac pathology is metabolic remodeling and loss of metabolic flexibility. Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuins (SIRT1 to SIRT7), with NAD+ dependent deacetylase activity. SIRT3 is expressed in high levels in healthy hearts but downregulated in the aged or diseased hearts. Experimental evidence shows that increasing SIRT3 levels or activity can ameliorate several cardiac pathologies. The primary deacetylation targets of SIRT3 are mitochondrial proteins, most of which are involved in energy metabolism. Thus, SIRT3 improves cardiac health by modulating cardiac energetics. In this review, we discuss the essential role of SIRT3 in regulating cardiac metabolism in the context of physiology and pathology. Specifically, we summarize the recent advancements that emphasize the critical role of SIRT3 as a master regulator of cardiac metabolism. We also present a comprehensive view of all known activators of SIRT3, and elaborate on their therapeutic potential to ameliorate energetic abnormalities in various cardiac pathologies.
Collapse
|
56
|
Bugga P, Alam MJ, Kumar R, Pal S, Chattopadyay N, Banerjee SK. Sirt3 ameliorates mitochondrial dysfunction and oxidative stress through regulating mitochondrial biogenesis and dynamics in cardiomyoblast. Cell Signal 2022; 94:110309. [PMID: 35304284 DOI: 10.1016/j.cellsig.2022.110309] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 12/22/2022]
Abstract
Sirtuins are the endogenously present anti-aging protein deacetylases that regulate the mitochondrial biogenesis and function. Especially Sirt3, a mitochondrial sirtuin, is well known for maintaining mitochondrial function and health. In the present study, we have explored the novel role of Sirt3 in mitochondrial biogenesis and shown the role of Sirt3 in mito-nuclear communication through AMPK-α in Sirt3 knockdown and Sirt3 overexpressed H9c2 cells. The study found that impaired mitochondrial function in Sirt3-knockdown H9c2 cells was associated with decreased expression of mitochondrial DNA encoded genes, reduced SOD2 expression and activity. The study also revealed that Sirt3 knockdown affects mitochondrial biogenesis and dynamics. To further confirm the role of Sirt3 on mitochondrial biogenesis and health, we did Sirt3 overexpression in H9c2 cells. Sirt3 overexpression enhanced the expression of mitochondrial DNA encoded genes, increased SOD2 activity and altered mitochondrial dynamics. Sirt3 overexpression also caused an increase in mitochondrial biogenesis gene and protein (PGC-1α and TFAM) expression. All these changes were confirmed with mitochondrial functional parameters like basal respiration, maximal respiratory capacity, spare respiratory capacity and ATP production. We found decreased mitochondrial function in Sirt3-knockdown H9c2 cells when compared to control H9c2 cells. Together our data conclude that Sirt3 regulates cardiac mitochondrial health and function through the Sirt3-AMPKα-PGC-1α axis.
Collapse
Affiliation(s)
- Paramesha Bugga
- Non-Communicable Diseases Group, Translational Health Science and Technology Institute (THSTI), Faridabad, 121001, Haryana, INDIA
| | - Md Jahangir Alam
- Non-Communicable Diseases Group, Translational Health Science and Technology Institute (THSTI), Faridabad, 121001, Haryana, INDIA; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Roshan Kumar
- Non-Communicable Diseases Group, Translational Health Science and Technology Institute (THSTI), Faridabad, 121001, Haryana, INDIA.
| | - Subhashis Pal
- Endocrinology Department, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naibedya Chattopadyay
- Endocrinology Department, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| | - Sanjay Kumar Banerjee
- Non-Communicable Diseases Group, Translational Health Science and Technology Institute (THSTI), Faridabad, 121001, Haryana, INDIA; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India.
| |
Collapse
|
57
|
Liu J, Tang M, Li T, Su Z, Zhu Z, Dou C, Liu Y, Pei H, Yang J, Ye H, Chen L. Honokiol Ameliorates Post-Myocardial Infarction Heart Failure Through Ucp3-Mediated Reactive Oxygen Species Inhibition. Front Pharmacol 2022; 13:811682. [PMID: 35264952 PMCID: PMC8899544 DOI: 10.3389/fphar.2022.811682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/20/2022] [Indexed: 01/31/2023] Open
Abstract
Post-myocardial infarction heart failure (post-MI HF) is one of the leading global causes of death, and current prevention and treatment methods still cannot avoid the increasing incidence. Honokiol (HK) has previously been reported to improve myocardial ischemia/reperfusion injury and reverse myocardial hypertrophy by activating Sirt1 and Sirt3. We suspect that HK may also have a therapeutic effect on post-MI HF. In this study, we aimed to investigate the efficacy and mechanism of HK in the treatment of post-MI HF. We found that HK inhibited myocardial reactive oxygen species (ROS) production, reduced myocardial fibrosis, and improved cardiac function in mice after MI. HK also reduced the abnormality of mitochondrial membrane potential (MMP) and apoptosis of cardiomyocytes caused by peroxide in neonatal cardiomyocytes. RNAseq results revealed that HK restored the transcriptome changes to a certain extent and significantly enhanced the expression of mitochondrial inner membrane uncoupling protein isoform 3 (Ucp3), a protein that inhibits the production of mitochondrial ROS, protects cardiomyocytes, and relieves heart failure after myocardial infarction (MI). In cardiomyocytes with impaired Ucp3 expression, HK cannot protect against the damage caused by peroxide. More importantly, in Ucp3 knockout mice, HK did not change the increase in the ROS level and cardiac function damage after MI. Taken together, our results suggest that HK can increase the expression of the cardioprotective protein Ucp3 and maintain MMP, thereby inhibiting the production of ROS after MI and ameliorating heart failure.
Collapse
Affiliation(s)
- Jianyu Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Li
- West China-Washington Mitochondria and Metabolism Center, Department of Anesthesiology, Laboratory of Anesthesiology and Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Zhengying Su
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Caixia Dou
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Heying Pei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lijuan Chen,
| |
Collapse
|
58
|
Cao M, Zhao Q, Sun X, Qian H, Lyu S, Chen R, Xia H, Yuan W. Sirtuin 3: Emerging therapeutic target for cardiovascular diseases. Free Radic Biol Med 2022; 180:63-74. [PMID: 35031448 DOI: 10.1016/j.freeradbiomed.2022.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/26/2022]
Abstract
Acetylation is one of the most important methods of modification that lead to a change in the function of proteins. In humans, metabolic enzymes commonly undergo acetylation, which regulates the activities of metabolic enzymes and metabolic pathways. Sirtuin 3 (SIRT3) is a prominent deacetylase that participates in mitochondrial metabolism, redox balance, and mitochondrial dynamics by regulating mitochondrial protein acetylation, thereby protecting mitochondria from damage. Normal mitochondrial function is essential for maintaining the metabolism and function of the heart. Therefore, mitochondrial dysfunction caused by SIRT3 consumption and defects leads to the development of a variety of cardiovascular diseases. A comprehensive understanding of the role of SIRT3 in cardiovascular disease is critical for developing new therapeutic strategies. Herein, we summarize the function of SIRT3 in mitochondria, the complex mechanisms mediating cardiovascular diseases, and the potential value of SIRT3 small-molecule agonists in future clinical treatments.
Collapse
Affiliation(s)
- Mengfei Cao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Qianru Zhao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Xia Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Han Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Shumei Lyu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Hao Xia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China.
| |
Collapse
|
59
|
Sheibani M, Azizi Y, Shayan M, Nezamoleslami S, Eslami F, Farjoo MH, Dehpour AR. Doxorubicin-Induced Cardiotoxicity: An Overview on Pre-clinical Therapeutic Approaches. Cardiovasc Toxicol 2022; 22:292-310. [DOI: 10.1007/s12012-022-09721-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/12/2022] [Indexed: 12/20/2022]
|
60
|
Videla LA, Marimán A, Ramos B, José Silva M, Del Campo A. Standpoints in mitochondrial dysfunction: Underlying mechanisms in search of therapeutic strategies. Mitochondrion 2022; 63:9-22. [PMID: 34990812 DOI: 10.1016/j.mito.2021.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction has been defined as a reduced efficiency of mitochondria to produce ATP given by a loss of mitochondrial membrane potential, alterations in the electron transport chain (ETC) function, with increase in reactive oxygen species (ROS) generation and decrease in oxygen consumption. During the last decades, mitochondrial dysfunction has been the focus of many researchers as a convergent point for the pathophysiology of several diseases. Numerous investigations have demonstrated that mitochondrial dysfunction is detrimental to cells, tissues and organisms, nevertheless, dysfunctional mitochondria can signal in a particular way in response to stress, a characteristic that may be useful to search for new therapeutic strategies with a common feature. The aim of this review addresses mitochondrial dysfunction and stress signaling as a promising target for future drug development.
Collapse
Affiliation(s)
- Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile.
| | - Andrea Marimán
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - Bastián Ramos
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - María José Silva
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile.
| |
Collapse
|
61
|
Tabrizi FB, Yarmohammadi F, Hayes AW, Karimi G. The modulation of SIRT1 and SIRT3 by natural compounds as a therapeutic target in doxorubicin-induced cardiotoxicity: A review. J Biochem Mol Toxicol 2021; 36:e22946. [PMID: 34747550 DOI: 10.1002/jbt.22946] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/28/2021] [Accepted: 10/22/2021] [Indexed: 12/23/2022]
Abstract
Doxorubicin (DOX) is a potent antitumor agent with a broad spectrum of activity; however, irreversible cardiotoxicity resulting from DOX treatment is a major issue that limits its therapeutic use. Sirtuins (SIRTs) play an essential role in several physiological and pathological processes including oxidative stress, apoptosis, and inflammation. It has been reported that SIRT1 and SIRT3 can act as a protective molecular against DOX-induced myocardial injury through targeting numerous signaling pathways. Several natural compounds (NCs), such as resveratrol, sesamin, and berberine, with antioxidative, anti-inflammation, and antiapoptotic effects were evaluated for their potential to suppress the cardiotoxicity induced by DOX via targeting SIRT1 and SIRT3. Numerous NCs exerted their therapeutic effects on DOX-mediated cardiac damage via targeting different signaling pathways, including SIRT1/LKB1/AMPK, SIRT1/PGC-1α, SIRT1/NLRP3, and SIRT3/FoxO. SIRT3 also ameliorates cardiotoxicity by enhancing mitochondrial fusion.
Collapse
Affiliation(s)
- Fatemeh B Tabrizi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA.,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
62
|
Yin Y, Shen H. Advances in Cardiotoxicity Induced by Altered Mitochondrial Dynamics and Mitophagy. Front Cardiovasc Med 2021; 8:739095. [PMID: 34616789 PMCID: PMC8488107 DOI: 10.3389/fcvm.2021.739095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 11/25/2022] Open
Abstract
Mitochondria are the most abundant organelles in cardiac cells, and are essential to maintain the normal cardiac function, which requires mitochondrial dynamics and mitophagy to ensure the stability of mitochondrial quantity and quality. When mitochondria are affected by continuous injury factors, the balance between mitochondrial dynamics and mitophagy is broken. Aging and damaged mitochondria cannot be completely removed in cardiac cells, resulting in energy supply disorder and accumulation of toxic substances in cardiac cells, resulting in cardiac damage and cardiotoxicity. This paper summarizes the specific underlying mechanisms by which various adverse factors interfere with mitochondrial dynamics and mitophagy to produce cardiotoxicity and emphasizes the crucial role of oxidative stress in mitophagy. This review aims to provide fresh ideas for the prevention and treatment of cardiotoxicity induced by altered mitochondrial dynamics and mitophagy.
Collapse
Affiliation(s)
- Yiyuan Yin
- Department of Emergency Medicine, ShengJing Hospital of China Medical University, Shenyang, China
| | - Haitao Shen
- Department of Emergency Medicine, ShengJing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
63
|
Kalyanaraman B. Reactive oxygen species, proinflammatory and immunosuppressive mediators induced in COVID-19: overlapping biology with cancer. RSC Chem Biol 2021; 2:1402-1414. [PMID: 34704045 PMCID: PMC8496060 DOI: 10.1039/d1cb00042j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
This review analyzes the published literature linking the different mechanisms focused on oxidative stress and inflammation that contribute to COVID-19 disease severity. The objective is to bring together potential proinflammatory mechanisms of COVID-19 pathogenesis and address mitigation strategies using naturally occurring compounds and FDA-approved drugs. Outstanding questions addressed include the following: What is the mechanistic basis for linking enhanced vulnerability in COVID-19 to increased oxidative damage and proinflammatory mediators (e.g., cytokines), especially in high-risk people? Can we repurpose anti-inflammatory and immunomodulatory agents to mitigate inflammation in COVID-19 patients? How does 2-deoxy-d-glucose function as an anti-COVID drug? COVID-19, cancer biology, and immunotherapy share many mechanistic similarities. Repurposing drugs that already have been FDA-approved for mitigating inflammation and immunosuppression in cancer may be a way to counteract disease severity, progression, and chronic inflammation in COVID-19. What are the long-term effects of reactive oxygen species-inducing immune cells and sustained inflammation in so-called long-haulers (long COVID) after recovery from COVID-19? Can we use mitochondria-targeted agents prophylactically to prevent inflammation and boost immunity in long-haulers? Addressing the oxidative chemical biology of COVID-19 and the mechanistic commonalities with cancer may provide new insights potentially leading to appropriate clinical trials and new treatments.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of Biophysics, Cancer Center, Center for Disease Prevention Research, Medical College of Wisconsin 8701 Watertown Plank Road Milwaukee WI 53226 USA
| |
Collapse
|
64
|
Arinno A, Maneechote C, Khuanjing T, Ongnok B, Prathumsap N, Chunchai T, Arunsak B, Kerdphoo S, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Cardioprotective effects of melatonin and metformin against doxorubicin-induced cardiotoxicity in rats are through preserving mitochondrial function and dynamics. Biochem Pharmacol 2021; 192:114743. [PMID: 34453902 DOI: 10.1016/j.bcp.2021.114743] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 01/23/2023]
Abstract
Doxorubicin (Dox) is widely used in chemotherapy regimens for several malignant conditions. Unfortunately, cumulative and irreversible cardiotoxicity of Dox is the most prominent adverse effect which limits its use. Several pharmacological interventions which exert antioxidant properties, including melatonin and metformin, have demonstrated beneficial effects against various cardiac pathological conditions. However, the exact molecular mechanisms underlying their cardioprotective effects are not completely understood. We hypothesized that treatment with either melatonin or metformin provides cardioprotection against Dox-induced cardiotoxicity through mitochondrial protection. Thirty-two male Wistar rats received 6 doses of either 0.9% normal saline solution (0.9% NSS, n = 8) or Dox (3 mg/kg, i.p., n = 24). The Dox-treated rats (n = 8/group) were co-treated with: 1) Vehicle (0.9% NSS), 2) Melatonin (10 mg/kg/day), and 3) Metformin (250 mg/kg/day) for 30 consecutive days via oral gavage. Following the treatment, left ventricular (LV) function, oxidative stress, inflammation, mitochondrial function, dynamics, biogenesis and bioenergetics, mitophagy, autophagy, and apoptosis were determined. Dox induced excessive oxidative stress, inflammation, autophagy, apoptosis, reduced mitochondrial function, dynamics balance, biogenesis, and bioenergetics leading to LV dysfunction. Treatment with either melatonin or metformin exerted equal measures of cardioprotection via reducing oxidative stress, inflammation, autophagy, apoptosis, and improved mitochondrial function, dynamics balance, biogenesis, and bioenergetics in the Dox-treated rats. Melatonin and metformin exerted both anti-cancer and cardioprotective properties, suggesting they have potential roles in concomitant therapy in cancer patients receiving Dox treatment.
Collapse
Affiliation(s)
- Apiwan Arinno
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Krekwit Shinlapawittayatorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
65
|
Chaanine AH. Metabolic Remodeling and Implicated Calcium and Signal Transduction Pathways in the Pathogenesis of Heart Failure. Int J Mol Sci 2021; 22:ijms221910579. [PMID: 34638917 PMCID: PMC8508915 DOI: 10.3390/ijms221910579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
The heart is an organ with high-energy demands in which the mitochondria are most abundant. They are considered the powerhouse of the cell and occupy a central role in cellular metabolism. The intermyofibrillar mitochondria constitute the majority of the three-mitochondrial subpopulations in the heart. They are also considered to be the most important in terms of their ability to participate in calcium and cellular signaling, which are critical for the regulation of mitochondrial function and adenosine triphosphate (ATP) production. This is because they are located in very close proximity with the endoplasmic reticulum (ER), and for the presence of tethering complexes enabling interorganelle crosstalk via calcium signaling. Calcium is an important second messenger that regulates mitochondrial function. It promotes ATP production and cellular survival under physiological changes in cardiac energetic demand. This is accomplished in concert with signaling pathways that regulate both calcium cycling and mitochondrial function. Perturbations in mitochondrial homeostasis and metabolic remodeling occupy a central role in the pathogenesis of heart failure. In this review we will discuss perturbations in ER-mitochondrial crosstalk and touch on important signaling pathways and molecular mechanisms involved in the dysregulation of calcium homeostasis and mitochondrial function in heart failure.
Collapse
Affiliation(s)
- Antoine H. Chaanine
- Department of Medicine, Heart and Vascular Institute, Tulane University, New Orleans, LA 70112, USA; ; Tel.: +1-(504)-988-1612
- Department of Physiology, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
66
|
Jakoube P, Cutano V, González-Morena JM, Keckesova Z. Mitochondrial Tumor Suppressors-The Energetic Enemies of Tumor Progression. Cancer Res 2021; 81:4652-4667. [PMID: 34183354 PMCID: PMC9397617 DOI: 10.1158/0008-5472.can-21-0518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023]
Abstract
Tumor suppressors represent a critical line of defense against tumorigenesis. Their mechanisms of action and the pathways they are involved in provide important insights into cancer progression, vulnerabilities, and treatment options. Although nuclear and cytosolic tumor suppressors have been extensively investigated, relatively little is known about tumor suppressors localized within the mitochondria. However, recent research has begun to uncover the roles of these important proteins in suppressing tumorigenesis. Here, we review this newly developing field and summarize available information on mitochondrial tumor suppressors.
Collapse
Affiliation(s)
- Pavel Jakoube
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Valentina Cutano
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Juan M. González-Morena
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Keckesova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.,Corresponding Author: Zuzana Keckesova, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 16000, Czech Republic. Phone: 420-2201-83584; E-mail:
| |
Collapse
|
67
|
Zhang Y, Wen P, Luo J, Ding H, Cao H, He W, Zen K, Zhou Y, Yang J, Jiang L. Sirtuin 3 regulates mitochondrial protein acetylation and metabolism in tubular epithelial cells during renal fibrosis. Cell Death Dis 2021; 12:847. [PMID: 34518519 PMCID: PMC8437958 DOI: 10.1038/s41419-021-04134-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/15/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
Proximal tubular epithelial cells (TECs) demand high energy and rely on mitochondrial oxidative phosphorylation as the main energy source. However, this is disturbed in renal fibrosis. Acetylation is an important post-translational modification for mitochondrial metabolism. The mitochondrial protein NAD+-dependent deacetylase sirtuin 3 (SIRT3) regulates mitochondrial metabolic function. Therefore, we aimed to identify the changes in the acetylome in tubules from fibrotic kidneys and determine their association with mitochondria. We found that decreased SIRT3 expression was accompanied by increased acetylation in mitochondria that have separated from TECs during the early phase of renal fibrosis. Sirt3 knockout mice were susceptible to hyper-acetylated mitochondrial proteins and to severe renal fibrosis. The activation of SIRT3 by honokiol ameliorated acetylation and prevented renal fibrosis. Analysis of the acetylome in separated tubules using LC-MS/MS showed that most kidney proteins were hyper-acetylated after unilateral ureteral obstruction. The increased acetylated proteins with 26.76% were mitochondrial proteins which were mapped to a broad range of mitochondrial pathways including fatty acid β-oxidation, the tricarboxylic acid cycle (TCA cycle), and oxidative phosphorylation. Pyruvate dehydrogenase E1α (PDHE1α), which is the primary link between glycolysis and the TCA cycle, was hyper-acetylated at lysine 385 in TECs after TGF-β1 stimulation and was regulated by SIRT3. Our findings showed that mitochondrial proteins involved in regulating energy metabolism were acetylated and targeted by SIRT3 in TECs. The deacetylation of PDHE1α by SIRT3 at lysine 385 plays a key role in metabolic reprogramming associated with renal fibrosis.
Collapse
Affiliation(s)
- Yu Zhang
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China
| | - Ping Wen
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China
| | - Jing Luo
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China
| | - Hao Ding
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China
| | - Hongdi Cao
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China
| | - Weichun He
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Nanjing, Jiangsu, 210093, China
| | - Yang Zhou
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China.
| | - Junwei Yang
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China.
| | - Lei Jiang
- Center for Kidney Disease, The second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210003, China.
| |
Collapse
|
68
|
Li D, Yang Y, Wang S, He X, Liu M, Bai B, Tian C, Sun R, Yu T, Chu X. Role of acetylation in doxorubicin-induced cardiotoxicity. Redox Biol 2021; 46:102089. [PMID: 34364220 PMCID: PMC8350499 DOI: 10.1016/j.redox.2021.102089] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
As a potent chemotherapeutic agent, doxorubicin (DOX) is widely used for the treatment of a variety of cancers However, its clinical utility is limited by dose-dependent cardiotoxicity, and pathogenesis has traditionally been attributed to the formation of reactive oxygen species (ROS). Accordingly, the prevention of DOX-induced cardiotoxicity is an indispensable goal to optimize therapeutic regimens and reduce morbidity. Acetylation is an emerging and important epigenetic modification regulated by histone deacetylases (HDACs) and histone acetyltransferases (HATs). Despite extensive studies of the molecular basis and biological functions of acetylation, the application of acetylation as a therapeutic target for cardiotoxicity is in the initial stage, and further studies are required to clarify the complex acetylation network and improve the clinical management of cardiotoxicity. In this review, we summarize the pivotal functions of HDACs and HATs in DOX-induced oxidative stress, the underlying mechanisms, the contributions of noncoding RNAs (ncRNAs) and exercise-mediated deacetylases to cardiotoxicity. Furthermore, we describe research progress related to several important SIRT activators and HDAC inhibitors with potential clinical value for chemotherapy and cardiotoxicity. Collectively, a comprehensive understanding of specific roles and recent developments of acetylation in doxorubicin-induced cardiotoxicity will provide a basis for improved treatment outcomes in cancer and cardiovascular diseases.
Collapse
Affiliation(s)
- Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao, 266071, China
| | - Shizhong Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Meixin Liu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Baochen Bai
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chao Tian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ruicong Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Basic Medicine School, Qingdao University, 38 Deng Zhou Road, Qingdao, 266021, China.
| | - Xianming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266071, China.
| |
Collapse
|
69
|
Rauf A, Olatunde A, Imran M, Alhumaydhi FA, Aljohani ASM, Khan SA, Uddin MS, Mitra S, Emran TB, Khayrullin M, Rebezov M, Kamal MA, Shariati MA. Honokiol: A review of its pharmacological potential and therapeutic insights. PHYTOMEDICINE 2021; 90:153647. [PMID: 34362632 DOI: 10.1016/j.phymed.2021.153647] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/17/2021] [Accepted: 06/28/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Honokiol is a pleiotropic compound which been isolated from Magnolia species such as Magnolia grandiflora and Magnolia dealbata. Magnolia species Magnolia grandiflora is used in traditional medicine for the treatment of various diseases. PURPOSE The objective of this review is to summarize the pharmacological potential and therapeutic insights of honokiol. STUDY DESIGN Honokiol has been specified as a novel alternative to treat various disorders such as liver cancer, neuroprotective, anti-spasmodic, antidepressant, anti-tumorigenic, antithrombotic, antimicrobial, analgesic properties and others. Therefore, this study designed to represent the in-depth therapeutic potential of honokiol. METHODS Literature searches in electronic databases, such as Web of Science, Science Direct, PubMed, Google Scholar, and Scopus, were performed using the keywords 'Honokiol', 'Health Benefits' and 'Therapeutic Insights' as the keywords for primary searches and secondary search terms were used as follows: 'Anticancer', 'Oxidative Stress', 'Neuroprotective', 'Antimicrobial', 'Cardioprotection', 'Hepatoprotective', 'Anti-inflammatory', 'Arthritis', 'Reproductive Disorders'. RESULTS This promising bioactive compound presented an wide range of therapeutic and biological activities which include liver cancer, neuroprotective, anti-spasmodic, antidepressant, anti-tumorigenic, antithrombotic, antimicrobial, analgesic properties, and others. Its pharmacokinetics has been established in experimental animals, while in humans, this is still speculative. Some of its mechanism for exhibiting its pharmacological effects includes apoptosis of diseased cells, reduction in the expression of defective proteins like P-glycoproteins, inhibition of oxidative stress, suppression of pro-inflammatory cytokines (TNF-α, IL-10 and IL-6), amelioration of impaired hepatic enzymes and reversal of morphological alterations, among others. CONCLUSION All these actions displayed by this novel compound could make it serve as a lead in the formulation of drugs with higher efficacy and negligible side effects utilized in the treatment of several human diseases.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, 23430, Khyber Pakhtunkhwa (KP), Pakistan.
| | - Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauchi, 740272, Nigeria
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, University of Lahore, Pakistan
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Shahid Ali Khan
- Department of Chemistry, University of Swabi, Swabi, Anbar, 23430, Khyber Pakhtunkhwa (KP), Pakistan
| | - Md Sahab Uddin
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka-1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong-4381, Bangladesh
| | - Mars Khayrullin
- K.G. Razumovsky Moscow State University of Technologies and Management (the First Cossack University), 109004, Moscow, Russian Federation
| | - Maksim Rebezov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russian Federation; V. M. Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, 109029, Moscow, Russian Federation.; Ural State Agrarian University, 620075 Yekaterinburg, Russian Federation
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| | - Mohammad Ali Shariati
- K.G. Razumovsky Moscow State University of Technologies and Management (the First Cossack University), 109004, Moscow, Russian Federation
| |
Collapse
|
70
|
Cheresh P, Kim SJ, Jablonski R, Watanabe S, Lu Z, Chi M, Helmin KA, Gius D, Budinger GRS, Kamp DW. SIRT3 Overexpression Ameliorates Asbestos-Induced Pulmonary Fibrosis, mt-DNA Damage, and Lung Fibrogenic Monocyte Recruitment. Int J Mol Sci 2021; 22:6856. [PMID: 34202229 PMCID: PMC8268084 DOI: 10.3390/ijms22136856] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/28/2023] Open
Abstract
Alveolar epithelial cell (AEC) mitochondrial (mt) DNA damage and fibrotic monocyte-derived alveolar macrophages (Mo-AMs) are implicated in the pathobiology of pulmonary fibrosis. We showed that sirtuin 3 (SIRT3), a mitochondrial protein regulating cell fate and aging, is deficient in the AECs of idiopathic pulmonary fibrosis (IPF) patients and that asbestos- and bleomycin-induced lung fibrosis is augmented in Sirt3 knockout (Sirt3-/-) mice associated with AEC mtDNA damage and intrinsic apoptosis. We determined whether whole body transgenic SIRT3 overexpression (Sirt3Tg) protects mice from asbestos-induced pulmonary fibrosis by mitigating lung mtDNA damage and Mo-AM recruitment. Crocidolite asbestos (100 µg/50 µL) or control was instilled intratracheally in C57Bl6 (Wild-Type) mice or Sirt3Tg mice, and at 21 d lung fibrosis (histology, fibrosis score, Sircol assay) and lung Mo-AMs (flow cytometry) were assessed. Compared to controls, Sirt3Tg mice were protected from asbestos-induced pulmonary fibrosis and had diminished lung mtDNA damage and Mo-AM recruitment. Further, pharmacologic SIRT3 inducers (i.e., resveratrol, viniferin, and honokiol) each diminish oxidant-induced AEC mtDNA damage in vitro and, in the case of honokiol, protection occurs in a SIRT3-dependent manner. We reason that SIRT3 preservation of AEC mtDNA is a novel therapeutic focus for managing patients with IPF and other types of pulmonary fibrosis.
Collapse
Affiliation(s)
- Paul Cheresh
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Seok-Jo Kim
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Renea Jablonski
- Section of Pulmonary and Critical Care, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Satoshi Watanabe
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Ziyan Lu
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Monica Chi
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Kathryn A. Helmin
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - David Gius
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - G. R. Scott Budinger
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - David W. Kamp
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| |
Collapse
|
71
|
Resveratrol Prevents Right Ventricle Dysfunction, Calcium Mishandling, and Energetic Failure via SIRT3 Stimulation in Pulmonary Arterial Hypertension. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9912434. [PMID: 34239697 PMCID: PMC8238598 DOI: 10.1155/2021/9912434] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary vessel remodeling; however, its severity and impact on survival depend on right ventricular (RV) failure. Resveratrol (RES), a polyphenol found in red wine, exhibits cardioprotective effects on RV dysfunction in PAH. However, most literature has focused on RES protective effect on lung vasculature; recent finding indicates that RES has a cardioprotective effect independent of pulmonary arterial pressure on RV dysfunction, although the underlying mechanism in RV has not been determined. Therefore, this study is aimed at evaluating sirtuin-3 (SIRT3) modulation by RES in RV using a monocrotaline- (MC-) induced PAH rat model. Myocyte function was evaluated by confocal microscopy as cell contractility, calcium signaling, and mitochondrial membrane potential (ΔΨm); cell energetics was assessed by high-resolution respirometry, and western blot and immunoprecipitation evaluated posttranslational modifications. PAH significantly affects mitochondrial function in RV; PAH is prone to mitochondrial permeability transition pore (mPTP) opening, thus decreasing the mitochondrial membrane potential. The compromised cellular energetics affects cardiomyocyte function by decreasing sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) activity and delaying myofilament unbinding, disrupting cell relaxation. RES partially protects mitochondrial integrity by deacetylating cyclophilin-D, a critical component of the mPTP, increasing SIRT3 expression and activity and preventing mPTP opening. The preserved energetic capability rescues cell relaxation by maintaining SERCA activity. Avoiding Ca2+ transient and cell contractility mismatch by preserving mitochondrial function describes, for the first time, impairment in excitation-contraction-energetics coupling in RV failure. These results highlight the importance of mitochondrial energetics and mPTP in PAH.
Collapse
|
72
|
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. [PMID: 34149423 PMCID: PMC8209419 DOI: 10.3389/fphar.2021.670479] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Today the pharmacological possibilities of treating cancer are expanding and as a result, life expectancy is increasing against the background of chemotherapy and supportive treatment. In the conditions of successful antitumor treatment, complications associated with its toxic effect on healthy tissues and organs began to come to the fore. Anthracycline cardiomyopathy was the first serious cardiovascular complication to draw the attention of oncologists and cardiologists around the world. Anthracycline drugs such as doxorubicin, epirubicin, idarubicin are still widely used in oncological practice to treat a wide range of solid and hematological malignancies. Doxorubicin-induced cardiomyopathy is closely associated with an increase in oxidative stress, as evidenced by reactive oxygen species (ROS) nduced damage such as lipid peroxidation, and decreased levels of antioxidants. Myofibrillar destruction and dysregulation of intracellular calcium are also important mechanisms, usually associated with doxorubicin-induced cardiotoxicity. Despite the abundance of data on various mechanisms involved in the implementation of doxorubicin-induced cardiotoxicity, a final understanding of the mechanism of the development of doxorubicin cardiomyopathy has not yet been formed. It poses the most significant challenges to the development of new methods of prevention and treatment, as well as to the unambiguous choice of a specific treatment regimen using the existing pharmacological tools. In order to resolve these issues new models that could reflect the development of the chemotherapy drugs effects are needed. In this review we have summarized and analyzed information on the main existing models of doxorubicin cardiomyopathy using small laboratory animals. In addition, this paper discusses further areas of research devoted to the development and validation of new improved models of doxorubicin cardiomyopathy suitable both for studying the mechanisms of its implementation and for the preclinical drugs effectiveness assessment.
Collapse
Affiliation(s)
- Ekaterina Yu Podyacheva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| | - Ekaterina A Kushnareva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| | - Andrei A Karpov
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| | - Yana G Toropova
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| |
Collapse
|
73
|
Tao RH, Kobayashi M, Yang Y, Kleinerman ES. Exercise Inhibits Doxorubicin-Induced Damage to Cardiac Vessels and Activation of Hippo/YAP-Mediated Apoptosis. Cancers (Basel) 2021; 13:cancers13112740. [PMID: 34205942 PMCID: PMC8198139 DOI: 10.3390/cancers13112740] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 01/16/2023] Open
Abstract
Dose-related cardiomyopathy is a major side effect following doxorubicin (Dox). To investigate whether exercise (Ex)-induced vasculogenesis plays a role in reducing Dox-induced cardiotoxicity, GFP+ bone marrow (BM) cells from GFP transgenic mice were transplanted into wild-type mice. Transplanted mice were treated with Dox, Ex, Dox+Ex, or control. We found Dox therapy resulted in decreased systolic and diastolic blood flow, decreased ejection fraction and fractional shortening, and decreased vascular endothelial cells and pericytes. These abnormalities were not seen in Dox+Ex hearts. Heart tissues from control-, Ex-, or Dox-treated mice showed a small number of GFP+ cells. By contrast, the Dox+Ex-treated hearts had a significant increase in GFP+ cells. Further analyses demonstrated these GFP+ BM cells had differentiated into vascular endothelial cells (GFP+CD31+) and pericytes (GFP+NG2+). Decreased cardiomyocytes were also seen in Dox-treated but not Dox+Ex-treated hearts. Ex induced an increase in GFP+c-Kit+ cells. However, these c-Kit+ BM stem cells had not differentiated into cardiomyocytes. Dox therapy induced phosphorylation of MST1/2, LATS1, and YAP; a decrease in total YAP; and cleavage of caspase-3 and PARP in the heart tissues. Dox+Ex prevented these effects. Our data demonstrated Dox-induced cardiotoxicity is mediated by vascular damage resulting in decreased cardiac blood flow and through activation of Hippo-YAP signaling resulting in cardiomyocyte apoptosis. Furthermore, Ex inhibited these effects by promoting migration of BM stem cells into the heart to repair the cardiac vessels damaged by Dox and through inhibiting Dox-induced Hippo-YAP signaling-mediated apoptosis. These data support the concept of using exercise as an intervention to decrease Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rong-Hua Tao
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| | - Masato Kobayashi
- School of Health Sciences, Institutes of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-0942, Japan;
| | - Yuanzheng Yang
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Eugenie S. Kleinerman
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| |
Collapse
|
74
|
Dewanjee S, Vallamkondu J, Kalra RS, Chakraborty P, Gangopadhyay M, Sahu R, Medala V, John A, Reddy PH, De Feo V, Kandimalla R. The Emerging Role of HDACs: Pathology and Therapeutic Targets in Diabetes Mellitus. Cells 2021; 10:1340. [PMID: 34071497 PMCID: PMC8228721 DOI: 10.3390/cells10061340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus (DM) is one of the principal manifestations of metabolic syndrome and its prevalence with modern lifestyle is increasing incessantly. Chronic hyperglycemia can induce several vascular complications that were referred to be the major cause of morbidity and mortality in DM. Although several therapeutic targets have been identified and accessed clinically, the imminent risk of DM and its prevalence are still ascending. Substantial pieces of evidence revealed that histone deacetylase (HDAC) isoforms can regulate various molecular activities in DM via epigenetic and post-translational regulation of several transcription factors. To date, 18 HDAC isoforms have been identified in mammals that were categorized into four different classes. Classes I, II, and IV are regarded as classical HDACs, which operate through a Zn-based mechanism. In contrast, class III HDACs or Sirtuins depend on nicotinamide adenine dinucleotide (NAD+) for their molecular activity. Functionally, most of the HDAC isoforms can regulate β cell fate, insulin release, insulin expression and signaling, and glucose metabolism. Moreover, the roles of HDAC members have been implicated in the regulation of oxidative stress, inflammation, apoptosis, fibrosis, and other pathological events, which substantially contribute to diabetes-related vascular dysfunctions. Therefore, HDACs could serve as the potential therapeutic target in DM towards developing novel intervention strategies. This review sheds light on the emerging role of HDACs/isoforms in diabetic pathophysiology and emphasized the scope of their targeting in DM for constituting novel interventional strategies for metabolic disorders/complications.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | | | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba 305 8565, Japan;
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | - Moumita Gangopadhyay
- School of Life Science and Biotechnology, ADAMAS University, Barasat, Kolkata 700126, West Bengal, India;
| | - Ranabir Sahu
- Department of Pharmaceutical Technology, University of North Bengal, Darjeeling 734013, West Bengal, India;
| | - Vijaykrishna Medala
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India;
| | - Albin John
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.J.); (P.H.R.)
| | - P. Hemachandra Reddy
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.J.); (P.H.R.)
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India;
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana, India
| |
Collapse
|
75
|
Leite JA, Ghirotto B, Targhetta VP, de Lima J, Câmara NOS. Sirtuins as pharmacological targets in neurodegenerative and neuropsychiatric disorders. Br J Pharmacol 2021; 179:1496-1511. [PMID: 34029375 DOI: 10.1111/bph.15570] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes that regulate several processes, such as transcription, cell proliferation, differentiation and development. HDACs are classified as either Zn2+ -dependent or NAD+ -dependent enzymes. Over the years, experimental and clinical evidence has demonstrated that HDAC modulation is a critical process in neurodegenerative and psychiatric disorders. Nevertheless, most of the studies have focused on the role of Zn2+ -dependent HDACs in the development of these diseases, although there is growing evidence showing that the NAD+ -dependent HDACs, known as sirtuins, are also very promising targets. This possibility has been strengthened by reports of decreased levels of NAD+ in CNS disorders, which can lead to alterations in sirtuin activation and therefore result in increased pathology. In this review, we discuss the role of sirtuins in neurodegenerative and neuropsychiatric disorders as well the possible rationale for them to be considered as pharmacological targets in future therapeutic interventions.
Collapse
Affiliation(s)
- Jefferson A Leite
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruno Ghirotto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vitor P Targhetta
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jean de Lima
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Niels O S Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Division of Nephrology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
76
|
Garg G, Singh AK, Singh S, Rizvi SI. Promising drug discovery strategies for sirtuin modulators: what lessons have we learnt? Expert Opin Drug Discov 2021; 16:915-927. [PMID: 33880981 DOI: 10.1080/17460441.2021.1915980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Sirtuins, NAD-dependent protein deacetylases, require NAD+ for enzymatic activity. Recent research has indicated that sirtuins have a key role in the regulation of gene expression, the cell cycle, apoptosis, neurodegeneration and several age-related diseases. In mammals, there are seven sirtuin isoforms (SIRT-1-7) that catalyze specific lysine substrate deacetylation. AREAS COVERED This review explains the current information on the structure, function and importance of sirtuin modulators. It also explores the possible therapeutic applications of sirtuin modulators and related small molecules in the context of various diseases. EXPERT OPINION Sirtuin's modulators open a new area of research for targeting pathological conditions. Sirtuin modulators, through their targeted function, may provide a possible tool for the amelioration of various diseases. However, the search of activators/inhibitors for sirtuins needs further research. The structural elucidation of sirtuins will create an understanding for the development of isoform-specific selective modulators. This could be a useful tool to determine the functions of individual sirtuins as potential therapeutic agents.
Collapse
Affiliation(s)
- Geetika Garg
- Department of Biochemistry, University of Allahabad, Allahabad, India.,Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Abhishek Kumar Singh
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India
| | - Sandeep Singh
- Department of Biochemistry, University of Allahabad, Allahabad, India
| | | |
Collapse
|
77
|
Sritharan S, Sivalingam N. A comprehensive review on time-tested anticancer drug doxorubicin. Life Sci 2021; 278:119527. [PMID: 33887349 DOI: 10.1016/j.lfs.2021.119527] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 03/31/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022]
Abstract
Doxorubicin or Adriamycin, is one of the most widely used chemotherapeutic drug for treating a myriad of cancers. It induces cell death through multiple intracellular targets: reactive oxygen species generation, DNA-adduct formation, topoisomerase II inhibition, histone eviction, Ca2+ and iron hemostasis regulation, and ceramide overproduction. Moreover, doxorubicin-treated dying cells undergo cellular modifications that enable neighboring dendritic cell activation and enhanced presentation of tumor antigen. In addition, doxorubicin also aids in the immune-mediated clearance of tumor cells. However, the development of chemoresistance and cardiotoxicity side effect has undermined its widespread applicability. Several formulations of doxorubicin and co-treatments with inhibitors, miRNAs, natural compounds and other chemotherapeutic drugs have been essential in reducing its dosage-dependent toxicity and combating the development of resistance. Further, more advanced research into the molecular mechanism of chemoresistance development would be vital in improving the overall survivability of clinical patients and in preventing cancer relapse.
Collapse
Affiliation(s)
- Sruthi Sritharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603 203 Chengalpattu District, Tamil Nadu, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603 203 Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
78
|
Christidi E, Brunham LR. Regulated cell death pathways in doxorubicin-induced cardiotoxicity. Cell Death Dis 2021; 12:339. [PMID: 33795647 PMCID: PMC8017015 DOI: 10.1038/s41419-021-03614-x] [Citation(s) in RCA: 323] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 02/01/2023]
Abstract
Doxorubicin is a chemotherapeutic drug used for the treatment of various malignancies; however, patients can experience cardiotoxic effects and this has limited the use of this potent drug. The mechanisms by which doxorubicin kills cardiomyocytes has been elusive and despite extensive research the exact mechanisms remain unknown. This review focuses on recent advances in our understanding of doxorubicin induced regulated cardiomyocyte death pathways including autophagy, ferroptosis, necroptosis, pyroptosis and apoptosis. Understanding the mechanisms by which doxorubicin leads to cardiomyocyte death may help identify novel therapeutic agents and lead to more targeted approaches to cardiotoxicity testing.
Collapse
Affiliation(s)
- Effimia Christidi
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC Canada
| | - Liam R. Brunham
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
79
|
Sanchez-Martin C, Menon D, Moroni E, Ferraro M, Masgras I, Elsey J, Arbiser JL, Colombo G, Rasola A. Honokiol Bis-Dichloroacetate Is a Selective Allosteric Inhibitor of the Mitochondrial Chaperone TRAP1. Antioxid Redox Signal 2021; 34:505-516. [PMID: 32438819 PMCID: PMC8020504 DOI: 10.1089/ars.2019.7972] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aims: TNF receptor-associated protein 1 (TRAP1), the mitochondrial paralog of the heat shock protein 90 (Hsp90) family of molecular chaperones, is required for neoplastic growth in several tumor cell models, where it inhibits succinate dehydrogenase (SDH) activity, thus favoring bioenergetic rewiring, maintenance of redox homeostasis, and orchestration of a hypoxia-inducible factor 1-alpha (HIF1α)-mediated pseudohypoxic program. Development of selective TRAP1 inhibitors is instrumental for targeted development of antineoplastic drugs, but it has been hampered up to now by the high degree of homology among catalytic pockets of Hsp90 family members. The vegetal derivative honokiol and its lipophilic bis-dichloroacetate ester, honokiol DCA (HDCA), are small-molecule compounds with antineoplastic activity. HDCA leads to oxidative stress and apoptosis in in vivo tumor models and displays an action that is functionally opposed to that of TRAP1, as it induces both SDH and the mitochondrial deacetylase sirtuin-3 (SIRT3), which further enhances SDH activity. We investigated whether HDCA could interact with TRAP1, inhibiting its chaperone function, and the effects of HDCA on tumor cells harboring TRAP1. Results: An allosteric binding site in TRAP1 is able to host HDCA, which inhibits TRAP1 but not Hsp90 ATPase activity. In neoplastic cells, HDCA reverts TRAP1-dependent downregulation of SDH, decreases proliferation rate, increases mitochondrial superoxide levels, and abolishes tumorigenic growth. Innovation: HDCA is a potential lead compound for the generation of antineoplastic approaches based on the allosteric inhibition of TRAP1 chaperone activity. Conclusions: We have identified a selective TRAP1 inhibitor that can be used to better dissect TRAP1 biochemical functions and to tailor novel tumor-targeting strategies.
Collapse
Affiliation(s)
| | - Daniela Menon
- Dipartimento di Scienze Biomediche, Università di Padova, Padova, Italy
| | - Elisabetta Moroni
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Milano, Italy
| | | | - Ionica Masgras
- Dipartimento di Scienze Biomediche, Università di Padova, Padova, Italy.,Istituto di Neuroscienze, CNR, Padova, Italy
| | - Justin Elsey
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jack L Arbiser
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Giorgio Colombo
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Milano, Italy.,Dipartimento di Chimica, Università di Pavia, Pavia, Italy
| | - Andrea Rasola
- Dipartimento di Scienze Biomediche, Università di Padova, Padova, Italy
| |
Collapse
|
80
|
Nicol M, Sadoune M, Polidano E, Launay JM, Samuel JL, Azibani F, Cohen-Solal A. Doxorubicin-induced and trastuzumab-induced cardiotoxicity in mice is not prevented by metoprolol. ESC Heart Fail 2021; 8:928-937. [PMID: 33529501 PMCID: PMC8006653 DOI: 10.1002/ehf2.13198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 12/04/2020] [Accepted: 12/28/2020] [Indexed: 01/01/2023] Open
Abstract
Aims Our objectives were to validate a murine model of chronic cardiotoxicity induced by Doxorubicin (Dox) and Trastuzumab (Trast) and to test the potential cardio‐protective effect of metoprolol. Methods and results Male C57Bl6 mice were intraperitoneally injected during 2 weeks with Dox (24 mg/kg) or saline, and then with Trast (10 mg/kg) or saline for two more weeks. Half of the mice received metoprolol (100 mg/kg). Cardiotoxicity was defined by a decline in left ventricular ejection fraction (LVEF) ≥ 10 points. At Day 42, Dox + Trast‐treated mice exhibited a 13‐points decline in LVEF (74 ± 2.6% vs. 87 ± 0.8% for control mice, P < 0.001) and a severe cardiac atrophy (heart weight: 105 ± 2.7 mg vs. 119 ± 3.9 mg for control mice, P < 0.01). This cardiac atrophy resulted from an excess of cardiac necrosis (assessed by plasma cardiac troponin I level: 3.2 ± 0.4 ng/L vs. 1.3 ± 0.06 ng/L for control mice, P < 0.01), an increase in apoptosis (caspase 3 activity showing a six‐fold increase for Dox + Trast‐treated mice vs. controls, P < 0.001), and cardiomyocyte atrophy (myocyte size: 0.67 ± 0.08 μm2 vs. 1.36 ± 0.10 μm2 for control mice, P < 0.001). In addition, Dox + Trast‐treated mice were shown to have an increased cardiac oxidative stress (164 ± 14 dihydroethidine‐marked nuclei per area vs. 56 ± 9.5 for control mice, P < 0.01) and increased cardiac fibrosis (the semi‐quantitative fibrosis score was three‐fold higher for Dox + Trast‐treated mice as compared with controls, P < 0.01). Metoprolol was not able to prevent either the decrease in LVEF or the severe cardiac atrophy, the cardiac necrosis, and the cardiac remodelling induced by chemotherapies. Conclusion A murine model of chronic cardiotoxicity induced by Dox and Trast was characterized by a decrease in cardiac function, a cardiac apoptosis and necrosis leading to cardiomyocyte atrophy. Metoprolol did not prevent this cardiotoxicity.
Collapse
Affiliation(s)
- Martin Nicol
- Cardiology Department, Lariboisiere Hospital, University of Paris, Paris, France.,Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Malha Sadoune
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Evelyne Polidano
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Jean Marie Launay
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Jane Lise Samuel
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Feriel Azibani
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Alain Cohen-Solal
- Cardiology Department, Lariboisiere Hospital, University of Paris, Paris, France.,Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| |
Collapse
|
81
|
Sirtuin 3 Downregulation in Mycobacterium tuberculosis-Infected Macrophages Reprograms Mitochondrial Metabolism and Promotes Cell Death. mBio 2021; 12:mBio.03140-20. [PMID: 33531400 PMCID: PMC7858060 DOI: 10.1128/mbio.03140-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tuberculosis, the disease caused by the bacterium M. tuberculosis, remains one of the top 10 causes of death worldwide. Macrophages, the first cells to encounter M. tuberculosis and critical for defense against infection, are hijacked by M. tuberculosis as a protected growth niche. M. tuberculosis-infected macrophages undergo metabolic reprogramming where key mitochondrial pathways are modulated, but the mechanisms driving this metabolic shift is unknown. Mycobacterium tuberculosis induces metabolic reprogramming in macrophages like the Warburg effect. This enhances antimicrobial performance at the expense of increased inflammation, which may promote a pathogen-permissive host environment. Since the NAD+-dependent protein deacetylase Sirtuin 3 (SIRT3) is an important regulator of mitochondrial metabolism and cellular redox homeostasis, we hypothesized that SIRT3 modulation mediates M. tuberculosis-induced metabolic reprogramming. Infection of immortalized and primary murine macrophages resulted in reduced levels of SIRT3 mRNA and protein and perturbation of SIRT3-regulated enzymes in the tricarboxylic acid cycle, electron transport chain, and glycolytic pathway. These changes were associated with increased reactive oxygen species and reduced antioxidant scavenging, thereby triggering mitochondrial stress and macrophage cell death. Relevance to tuberculosis disease in vivo was indicated by greater bacterial burden and immune pathology in M. tuberculosis-infected Sirt3−/− mice. CD11b+ lung leukocytes isolated from infected Sirt3−/− mice showed decreased levels of enzymes involved in central mitochondrial metabolic pathways, along with increased reactive oxygen species. Bacterial burden was also greater in lungs of LysMcreSirt3L2/L2 mice, demonstrating the importance of macrophage-specific SIRT3 after infection. These results support the model of SIRT3 as a major upstream regulatory factor, leading to metabolic reprogramming in macrophages by M. tuberculosis.
Collapse
|
82
|
Li Q, Cheng JC, Jiang Q, Lee WY. Role of sirtuins in bone biology: Potential implications for novel therapeutic strategies for osteoporosis. Aging Cell 2021; 20:e13301. [PMID: 33393735 PMCID: PMC7884050 DOI: 10.1111/acel.13301] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022] Open
Abstract
The decline in bone mass and bone strength and musculoskeletal problems associated with aging constitute a major challenge for affected individuals and the healthcare system globally. Sirtuins 1-7 (SIRT1-SIRT7) are a family of nicotinamide adenine dinucleotide-dependent deacetylases with remarkable abilities to promote longevity and counteract age-related diseases. Sirtuin knockout and transgenic models have provided novel insights into the function and signaling of these proteins in bone homeostasis. Studies have revealed that sirtuins play a critical role in normal skeletal development and homeostasis through their direct action on bone cells and that their dysregulation might contribute to different bone diseases. Preclinical studies have demonstrated that mice treated with sirtuin agonists show protection against age-related, postmenopausal, and immobilization-induced osteoporosis. These findings suggest that sirtuins could be potential targets for the modulation of the imbalance in bone remodeling and treatment of osteoporosis and other bone disorders. The aim of this review was to provide a comprehensive updated review of the current knowledge on sirtuin biology, focusing specifically on their roles in bone homeostasis and osteoporosis, and potential pharmacological interventions targeting sirtuins for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Qiangqiang Li
- SH Ho Scoliosis Research LaboratoryDepartment of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing UniversityThe Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Jack Chun‐yiu Cheng
- SH Ho Scoliosis Research LaboratoryDepartment of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing UniversityThe Chinese University of Hong KongHong Kong SARChina
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive SurgeryDrum Tower Hospital affiliated to Medical School of Nanjing UniversityNanjingChina
| | - Wayne Yuk‐wai Lee
- SH Ho Scoliosis Research LaboratoryDepartment of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing UniversityThe Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
83
|
Mitochondrial Sirtuins and Doxorubicin-induced Cardiotoxicity. Cardiovasc Toxicol 2021; 21:179-191. [PMID: 33438065 DOI: 10.1007/s12012-020-09626-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOX) is the most effective and extensively used treatment for many tumors. However, its clinical use is hampered by its cardiotoxicity. DOX-induced mitochondrial dysfunction, which causes reactive oxygen species (ROS) generation, cardiomyocyte death, bioenergetic failure, and decreased cardiac function, is a very important mechanism of cardiotoxicity. These cellular processes are all linked by mitochondrial sirtuins (SIRT3-SIRT4). Mitochondrial sirtuins preserve mitochondrial function by increasing mitochondrial metabolism, inhibiting ROS generation by activating the antioxidant enzyme manganese-dependent superoxide dismutase (MnSOD), decreasing apoptosis by activating the forkhead homeobox type O (FOXO) and P53 pathways, and increasing autophagy through AMP-activated protein kinase (AMPK)/mTOR signaling. Thus, sirtuins function at the control point of many mechanisms involved in DOX-induced cardiotoxicity. In this review, we focus on the role of mitochondrial sirtuins in mitochondrial biology and DOX-induced cardiotoxicity. A further aim is to highlight other mitochondrial processes, such as autophagy (mitophagy) and mitochondrial quality control (MQC), for which the effect of mitochondrial sirtuins on cardiotoxicity is unknown.
Collapse
|
84
|
Shen Y, Wu Q, Shi J, Zhou S. Regulation of SIRT3 on mitochondrial functions and oxidative stress in Parkinson's disease. Biomed Pharmacother 2020; 132:110928. [PMID: 33128944 DOI: 10.1016/j.biopha.2020.110928] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Sirtuin-3 (SIRT3) is a NAD+-dependent protein deacetylase that is located in mitochondria, regulating mitochondrial proteins and maintaining cellular antioxidant status. Increasing evidence demonstrates that SIRT3 plays a role in degenerative disorders including Parkinson's disease (PD), which is a devastating nervous system disease currently with no effective treatments available. Although the etiology of PD is still largely ambiguous, substantial evidence indicates that mitochondrial dysfunction and oxidative stress play major roles in the pathogenesis of PD. The imbalance of reactive oxygen species (ROS) production and detoxification leads to oxidative stress that can accelerate the progression of PD. By causing conformational changes in the deacetylated proteins SIRT3 modulates the activities and biological functions of a variety of proteins involved in mitochondrial antioxidant defense and various mitochondrial functions. Increasingly more studies have suggested that upregulation of SIRT3 confers beneficial effect on neuroprotection in various PD models. This review discusses the mechanism by which SIRT3 regulates intracellular oxidative status and mitochondrial function with an emphasis in discussing in detail the regulation of SIRT3 on each component of the five complexes of the mitochondrial respiratory chain and mitochondrial antioxidant defense, as well as the pharmacological regulation of SIRT3 in light of therapeutic strategies for PD.
Collapse
Affiliation(s)
- Yanhua Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China.
| |
Collapse
|
85
|
Ni J, Liu Y, Wang K, Wu M, Kang L, Sha D, Xu B, Gu R. Trophoblast Stem-Cell-Derived Exosomes Improve Doxorubicin-Induced Dilated Cardiomyopathy by Modulating the let-7i/YAP Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:948-956. [PMID: 33294288 PMCID: PMC7680701 DOI: 10.1016/j.omtn.2020.10.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/11/2020] [Indexed: 11/25/2022]
Abstract
Trophoblast stem cells (TSCs) have been confirmed to play a cardioprotective role in heart failure. However, whether TSC-derived exosomes (TSC-exos) can protect against cardiac injury remains unclear. In the present study, TSC-exos were isolated from the supernatant of TSCs using the ultracentrifugation method and characterized by transmission electron microscopy and western blotting. Utilizing the public Gene Expression Omnibus (GEO) database, we found that let-7i and Yes-associated protein 1 (YAP) could participate in the development of heart failure. In vitro, AC16 cardiomyocytes subjected to doxorubicin (DOX) were treated with TSC-exos or let-7i mimic. Flow cytometry showed that TSC-exos and let-7i both decreased cardiomyocyte apoptosis. In vivo, mice that were intraperitoneally injected into DOX received either PBS, TSC-exos, or AAV9-let7iup for let-7i overexpression. Mice receiving TSC-exos and AAV9-let7iup showed improved cardiac function and decreased inflammatory responses, accompanied by downregulated YAP signaling. Mechanistically, TSC-exos could transfer let-7i to cardiomyocytes and silence the YAP signaling pathway. In conclusion, TSC-exos could alleviate DOX-induced cardiac injury via the let-7i/YAP pathway, which sheds new light on the application of TSC-exos as a potential therapeutic tool for heart failure.
Collapse
Affiliation(s)
- Jie Ni
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 21008, P.R. China.,Department of General Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, P.R. China
| | - Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Kun Wang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 21008, P.R. China
| | - Mingyue Wu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 21008, P.R. China
| | - Lina Kang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 21008, P.R. China
| | - Dujuan Sha
- Department of General Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, P.R. China
| | - Biao Xu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 21008, P.R. China
| | - Rong Gu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 21008, P.R. China
| |
Collapse
|
86
|
Xia B, Tong Y, Xia C, Chen C, Shan X. α-Cyperone Confers Antidepressant-Like Effects in Mice via Neuroplasticity Enhancement by SIRT3/ROS Mediated NLRP3 Inflammasome Deactivation. Front Pharmacol 2020; 11:577062. [PMID: 33132912 PMCID: PMC7579414 DOI: 10.3389/fphar.2020.577062] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022] Open
Abstract
α-Cyperone (Cy) is a major active compound of Cyperus rotundus that has various pharmacological activities. But whether Cy possesses antidepressant effect is unknown. In this study, we exposed mice to chronic unpredictable mild stress (CUMS) with or without intervention with Cy. Our results showed that Cy significantly improved the depressive phenotypes in sucrose preference test, tail suspension test and forced swimming test. Meanwhile, increased SIRT3 expression, reduced ROS production and activated NF-κB signal were detected in the hippocampus of mice. NLRP3 inflammasome related proteins including NLRP3, ASC, Caspase-1, IL-1β, IL-18 and GSDMD-N were downregulated after Cy administration. Synaptic proteins including Synapsin-1 and PSD-95 and dendritic spine density were improved after Cy treatment. Moreover, the protective effects of Cy in CUMS mice were compromised when co-administrated with SIRT3 inhibitor 3-TYP. Taken together, these findings suggested that Cy has therapeutic potential for treating depression and that this antidepressant effect may be attributed to SIRT3 stimulated neuroplasticity enhancement by suppressing NLRP3 inflammasome.
Collapse
Affiliation(s)
- Baomei Xia
- Faculty of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing, China.,Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Yue Tong
- School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Taizhou, China
| | - Changbo Xia
- School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Taizhou, China
| | - Chang Chen
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Shan
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, China
| |
Collapse
|
87
|
Sodium nitrate co-supplementation does not exacerbate low dose metronomic doxorubicin-induced cachexia in healthy mice. Sci Rep 2020; 10:15044. [PMID: 32973229 PMCID: PMC7518269 DOI: 10.1038/s41598-020-71974-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
The purpose of this study was to determine whether (1) sodium nitrate (SN) treatment progressed or alleviated doxorubicin (DOX)-induced cachexia and muscle wasting; and (2) if a more-clinically relevant low-dose metronomic (LDM) DOX treatment regimen compared to the high dosage bolus commonly used in animal research, was sufficient to induce cachexia in mice. Six-week old male Balb/C mice (n = 16) were treated with three intraperitoneal injections of either vehicle (0.9% NaCl; VEH) or DOX (4 mg/kg) over one week. To test the hypothesis that sodium nitrate treatment could protect against DOX-induced symptomology, a group of mice (n = 8) were treated with 1 mM NaNO3 in drinking water during DOX (4 mg/kg) treatment (DOX + SN). Body composition indices were assessed using echoMRI scanning, whilst physical and metabolic activity were assessed via indirect calorimetry, before and after the treatment regimen. Skeletal and cardiac muscles were excised to investigate histological and molecular parameters. LDM DOX treatment induced cachexia with significant impacts on both body and lean mass, and fatigue/malaise (i.e. it reduced voluntary wheel running and energy expenditure) that was associated with oxidative/nitrostative stress sufficient to induce the molecular cytotoxic stress regulator, nuclear factor erythroid-2-related factor 2 (NRF-2). SN co-treatment afforded no therapeutic potential, nor did it promote the wasting of lean tissue. Our data re-affirm a cardioprotective effect for SN against DOX-induced collagen deposition. In our mouse model, SN protected against LDM DOX-induced cardiac fibrosis but had no effect on cachexia at the conclusion of the regimen.
Collapse
|
88
|
Kerr M, Miller JJ, Thapa D, Stiewe S, Timm KN, Aparicio CNM, Scott I, Tyler DJ, Heather LC. Rescue of myocardial energetic dysfunction in diabetes through the correction of mitochondrial hyperacetylation by honokiol. JCI Insight 2020; 5:140326. [PMID: 32879143 PMCID: PMC7526448 DOI: 10.1172/jci.insight.140326] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/16/2020] [Indexed: 11/22/2022] Open
Abstract
Cardiac energetic dysfunction has been reported in patients with type 2 diabetes (T2D) and is an independent predictor of mortality. Identification of the mechanisms driving mitochondrial dysfunction, and therapeutic strategies to rescue these modifications, will improve myocardial energetics in T2D. We demonstrate using 31P-magnetic resonance spectroscopy (31P-MRS) that decreased cardiac ATP and phosphocreatine (PCr) concentrations occurred before contractile dysfunction or a reduction in PCr/ATP ratio in T2D. Real-time mitochondrial ATP synthesis rates and state 3 respiration rates were similarly depressed in T2D, implicating dysfunctional mitochondrial energy production. Driving this energetic dysfunction in T2D was an increase in mitochondrial protein acetylation, and increased ex vivo acetylation was shown to proportionally decrease mitochondrial respiration rates. Treating T2D rats in vivo with the mitochondrial deacetylase SIRT3 activator honokiol reversed the hyperacetylation of mitochondrial proteins and restored mitochondrial respiration rates to control levels. Using 13C-hyperpolarized MRS, respiration with different substrates, and enzyme assays, we localized this improvement to increased glutamate dehydrogenase activity. Finally, honokiol treatment increased ATP and PCr concentrations and increased total ATP synthesis flux in the T2D heart. In conclusion, hyperacetylation drives energetic dysfunction in T2D, and reversing acetylation with the SIRT3 activator honokiol rescued myocardial and mitochondrial energetics in T2D. Pharmacologically targeting mitochondrial acetylation provides a mechanism to rescue impaired myocardial energy generation in type 2 diabetes.
Collapse
Affiliation(s)
- Matthew Kerr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Jack J Miller
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, United Kingdom.,Department of Physics, University of Oxford, Oxford, United Kingdom
| | - Dharendra Thapa
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sophie Stiewe
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kerstin N Timm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Iain Scott
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
89
|
Zhou ZD, Tan EK. Oxidized nicotinamide adenine dinucleotide-dependent mitochondrial deacetylase sirtuin-3 as a potential therapeutic target of Parkinson's disease. Ageing Res Rev 2020; 62:101107. [PMID: 32535274 DOI: 10.1016/j.arr.2020.101107] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/18/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
Abstract
Mitochondrial impairment is associated with progressive dopamine (DA) neuron degeneration in Parkinson's disease (PD). Recent findings highlight that Sirtuin-3 (SIRT3), a mitochondrial protein, is an oxidized nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase and a key modulator in maintaining integrity and functions of mitochondria. SIRT3 plays vital roles in regulation of mitochondrial functions, including mitochondrial ATP generation and energy metabolism, anti-oxidant defense, and cell death and proliferation. SIRT3 can deacetylate the transcriptional factors and crosstalk with different signaling pathways to cooperatively modulate mitochondrial functions and regulate defensive mitochondrial quality control (QC) systems. Down-regulated NAD+ level and decreased SIRT3 activity are related to aging process and has been pathologically linked to PD pathogenesis. Further, SIRT3 can bind and deacetylate PTEN-induced kinase 1 (PINK1) and PD protein 2 E3 ubiquitin protein ligase (Parkin) to facilitate mitophagy. Leucine Rich Repeat Kinase 2 (LRRK2)-G2019S mutation in PD is linked to SIRT3 impairment. Furthermore, SIRT3 is inversely associated with α-synuclein aggregation and DA neuron degeneration in PD. SIRT3 chemical activators and NAD+ precursors can up-regulate SIRT3 activity to protect against DA neuron degeneration in PD models. Taken together, SIRT3 is a promising PD therapeutic target and studies of SIRT3 functional modulators with neuroprotective capability will be of clinical interest.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, 308433, Singapore; Duke-NUS Graduate Medical School, 8 College Road, 169857, Singapore.
| | - Eng King Tan
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, 308433, Singapore; Department of Neurology, Singapore General Hospital, Outram Road, 169608, Singapore; Duke-NUS Graduate Medical School, 8 College Road, 169857, Singapore.
| |
Collapse
|
90
|
Caballero EP, Mariz-Ponte N, Rigazio CS, Santamaría MH, Corral RS. Honokiol attenuates oxidative stress-dependent heart dysfunction in chronic Chagas disease by targeting AMPK / NFE2L2 / SIRT3 signaling pathway. Free Radic Biol Med 2020; 156:113-124. [PMID: 32540353 DOI: 10.1016/j.freeradbiomed.2020.05.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/11/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Eugenia Pérez Caballero
- Laboratorio de Biología Experimental, Centro de Estudios Metabólicos, Santander, 39005, Spain
| | - Nilo Mariz-Ponte
- Instituto de Investigação Biomédica, Universidade de Coimbra, Coimbra, 3004517, Portugal
| | - Cristina S Rigazio
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP, CONICET-GCBA), Servicio de Parasitología-Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", Buenos Aires, 1425, Argentina
| | - Miguel H Santamaría
- Laboratorio de Biología Experimental, Centro de Estudios Metabólicos, Santander, 39005, Spain
| | - Ricardo S Corral
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP, CONICET-GCBA), Servicio de Parasitología-Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", Buenos Aires, 1425, Argentina.
| |
Collapse
|
91
|
Yang SR, Hsu WH, Wu CY, Shang HS, Liu FC, Chen A, Hua KF, Ka SM. Accelerated, severe lupus nephritis benefits from treatment with honokiol by immunoregulation and differentially regulating NF-κB/NLRP3 inflammasome and sirtuin 1/autophagy axis. FASEB J 2020; 34:13284-13299. [PMID: 32813287 DOI: 10.1096/fj.202001326r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022]
Abstract
Using honokiol (HNK), a major anti-inflammatory bioactive compound in Magnolia officinalis, we show a potent therapeutic outcome against an accelerated, severe form of lupus nephritis (ASLN). The latter may follow infectious insults that act as environmental triggers in the patients. In the current study, an ASLN model in NZB/W F1 mice was treated with HNK by daily gavage after onset of the disease. We show that HNK ameliorated the ASLN by improving renal function, albuminuria, and renal pathology, especially reducing cellular crescents, neutrophil influx, fibrinoid necrosis in glomeruli, and glomerulonephritis activity scores. Meanwhile, HNK differentially regulated T cell functions, reduced serum anti-dsDNA autoantibodies, and inhibited NLRP3 inflammasome activation in the mice. The latter involved: (a) suppressed production of reactive oxygen species and NF-κB activation-mediated priming signal of the inflammasome, (b) reduced mitochondrial damage, and (c) enhanced sirtuin 1 (SIRT1)/autophagy axis activation. In conclusion, HNK represents a new drug candidate for acute, severe episodes of LN capable of alleviating renal lesions in ASLN mice by negatively regulating T cell functions and by enhancing SIRT1/autophagy axis-lessened NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Shin-Ruen Yang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wan-Han Hsu
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Yao Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ann Chen
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shuk-Man Ka
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Aerospace and Undersea Medicine, Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
92
|
Zhang J, Xiang H, Liu J, Chen Y, He RR, Liu B. Mitochondrial Sirtuin 3: New emerging biological function and therapeutic target. Theranostics 2020; 10:8315-8342. [PMID: 32724473 PMCID: PMC7381741 DOI: 10.7150/thno.45922] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/08/2020] [Indexed: 02/05/2023] Open
Abstract
Sirtuin 3 (SIRT3) is one of the most prominent deacetylases that can regulate acetylation levels in mitochondria, which are essential for eukaryotic life and inextricably linked to the metabolism of multiple organs. Hitherto, SIRT3 has been substantiated to be involved in almost all aspects of mitochondrial metabolism and homeostasis, protecting mitochondria from a variety of damage. Accumulating evidence has recently documented that SIRT3 is associated with many types of human diseases, including age-related diseases, cancer, heart disease and metabolic diseases, indicating that SIRT3 can be a potential therapeutic target. Here we focus on summarizing the intricate mechanisms of SIRT3 in human diseases, and recent notable advances in the field of small-molecule activators or inhibitors targeting SIRT3 as well as their potential therapeutic applications for future drug discovery.
Collapse
|
93
|
Sirtuin 3, Endothelial Metabolic Reprogramming, and Heart Failure With Preserved Ejection Fraction. J Cardiovasc Pharmacol 2020; 74:315-323. [PMID: 31425381 DOI: 10.1097/fjc.0000000000000719] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The incidences of heart failure with preserved ejection fraction (HFpEF) are increased in aged populations as well as diabetes and hypertension. Coronary microvascular dysfunction has contributed to the development of HFpEF. Endothelial cells (ECs) depend on glycolysis rather than oxidative phosphorylation for generating adenosine triphosphate to maintain vascular homeostasis. Glycolytic metabolism has a critical role in the process of angiogenesis, because ECs rely on the energy produced predominantly from glycolysis for migration and proliferation. Sirtuin 3 (SIRT3) is found predominantly in mitochondria and its expression declines progressively with aging, diabetes, obesity, and hypertension. Emerging evidence indicates that endothelial SIRT3 regulates a metabolic switch between glycolysis and mitochondrial respiration. SIRT3 deficiency in EC resulted in a significant decrease in glycolysis, whereas, it exhibited higher mitochondrial respiration and more prominent production of reactive oxygen species. SIRT3 deficiency also displayed striking increases in acetylation of p53, EC apoptosis, and senescence. Impairment of SIRT3-mediated EC metabolism may lead to a disruption of EC/pericyte/cardiomyocyte communications and coronary microvascular rarefaction, which promotes cardiomyocyte hypoxia, Titin-based cardiomyocyte stiffness, and myocardial fibrosis, thus leading to a diastolic dysfunction and HFpEF. This review summarizes current knowledge of SIRT3 in EC metabolic reprograming, EC/pericyte interactions, coronary microvascular dysfunction, and HFpEF.
Collapse
|
94
|
Lovy A, Ahumada-Castro U, Bustos G, Farias P, Gonzalez-Billault C, Molgó J, Cardenas C. Concerted Action of AMPK and Sirtuin-1 Induces Mitochondrial Fragmentation Upon Inhibition of Ca 2+ Transfer to Mitochondria. Front Cell Dev Biol 2020; 8:378. [PMID: 32523953 PMCID: PMC7261923 DOI: 10.3389/fcell.2020.00378] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/27/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are highly dynamic organelles constantly undergoing fusion and fission. Ca2+ regulates many aspects of mitochondrial physiology by modulating the activity of several mitochondrial proteins. We previously showed that inhibition of constitutive IP3R-mediated Ca2+ transfer to the mitochondria leads to a metabolic cellular stress and eventually cell death. Here, we show that the decline of mitochondrial function generated by a lack of Ca2+ transfer induces a DRP-1 independent mitochondrial fragmentation that at an early time is mediated by an increase in the NAD+/NADH ratio and activation of SIRT1. Subsequently, AMPK predominates and drives the fragmentation. SIRT1 activation leads to the deacetylation of cortactin, favoring actin polymerization, and mitochondrial fragmentation. Knockdown of cortactin or inhibition of actin polymerization prevents fragmentation. These data reveal SIRT1 as a new player in the regulation of mitochondrial fragmentation induced by metabolic/bioenergetic stress through regulating the actin cytoskeleton.
Collapse
Affiliation(s)
- Alenka Lovy
- Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, United States.,Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Galdo Bustos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Paula Farias
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Christian Gonzalez-Billault
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile
| | - Jordi Molgó
- Université Paris-Saclay, CEA, Institut des Sciences du Vivant Frédéric Joliot, ERL CNRS n° 9004, Département Médicaments et Technologies pour la Santé, Service d'Ingénierie Moléculaire pour la Santé (SIMoS), Gif-sur-Yvette, France
| | - Cesar Cardenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, United States.,Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
95
|
Sinha S, Sharma S, Vora J, Shrivastava N. Emerging role of sirtuins in breast cancer metastasis and multidrug resistance: Implication for novel therapeutic strategies targeting sirtuins. Pharmacol Res 2020; 158:104880. [PMID: 32442721 DOI: 10.1016/j.phrs.2020.104880] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
Sirtuins (SIRTs), a class III histone deacetylases (HDACs) that require NAD+ as a cofactor and include SIRT1-7 proteins in mammals. Accumulative evidence has established that every sirtuin possesses exclusive and poised biology, implicating their role in the regulation of multifaceted biological functions leading to breast cancer initiation, progression, and metastasis. This article provides an outline of recent developments in the role of sirtuins in breast cancer metastasis and development of multidrug resistance (MDR). In addition, we have also highlighted the impending prospects of targeting SIRTs to overcome MDR to bring advancement in breast cancer management. Further, this review will focus on strategies for improving the activity and efficacy of existing cancer therapeutics by combining (adjuvant treatment/therapy) them with sirtuin inhibitors/modulators. All available as well as newly discovered synthetic and dietary sirtuin inhibitors, activators/modulators have been extensively reviewed and compiled to provide a rationale for targeting sirtuins. Further, we discuss their potential in developing future therapeutics against sirtuins proposing their use along with conventional chemotherapeutics to overcome the problem of breast cancer metastasis and MDR.
Collapse
Affiliation(s)
- Sonam Sinha
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Sonal Sharma
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India
| | - Jaykant Vora
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Neeta Shrivastava
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India.
| |
Collapse
|
96
|
Irigenin treatment alleviates doxorubicin (DOX)-induced cardiotoxicity by suppressing apoptosis, inflammation and oxidative stress via the increase of miR-425. Biomed Pharmacother 2020; 125:109784. [PMID: 32092815 DOI: 10.1016/j.biopha.2019.109784] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/27/2019] [Accepted: 12/08/2019] [Indexed: 01/22/2023] Open
|
97
|
Yuan Y, Zhou X, Wang Y, Wang Y, Teng X, Wang S. Cardiovascular Modulating Effects of Magnolol and Honokiol, Two Polyphenolic Compounds from Traditional Chinese Medicine-Magnolia Officinalis. Curr Drug Targets 2020; 21:559-572. [PMID: 31749425 DOI: 10.2174/1389450120666191024175727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/09/2023]
Abstract
Honokiol and its isomer magnolol are poly-phenolic compounds isolated from the Magnolia officinalis that exert cardiovascular modulating effects via a variety of mechanisms. They are used as blood-quickening and stasis-dispelling agents in Traditional Chinese Medicine and confirmed to have therapeutic potential in atherosclerosis, thrombosis, hypertension, and cardiac hypertrophy. This comprehensive review summarizes the current data regarding the cardioprotective mechanisms of those compounds and identifies areas for further research.
Collapse
Affiliation(s)
- Yuan Yuan
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaocui Zhou
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Yuanyuan Wang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Yan Wang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Xiangyan Teng
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Shuaiyu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
98
|
Jo H, Park Y, Kim T, Kim J, Lee JS, Kim SY, Chung JI, Ko HY, Pyun JC, Kim KS, Lee M, Yun M. Modulation of SIRT3 expression through CDK4/6 enhances the anti-cancer effect of sorafenib in hepatocellular carcinoma cells. BMC Cancer 2020; 20:332. [PMID: 32306906 PMCID: PMC7168998 DOI: 10.1186/s12885-020-06822-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the leading cause of cancer-related deaths worldwide. The only drug currently approved for clinical use in the treatment of advanced HCC is sorafenib. However, many patients with HCC show reduced sensitivity to sorafenib during treatment. SIRT3, a member of the mammalian sirtuin family, is a tumor suppressor in certain tumor types. However, only few studies have investigated the effects of SIRT3 on tumor prognosis and sorafenib sensitivity in patients with HCC. Here, we aimed to investigate the correlation between SIRT3 expression and glucose metabolism and proliferation in HCC and discover effective compounds that increase endogenous SIRT3 modulation effect of sorafenib. Methods To determine the correlation between SIRT3 and glucose related proteins, immunostaining was performed with liver cancer tissue using various antibodies. To investigate whether the expression of SIRT3 in HCC is related to the resistance to sorafenib, we treated sorafenib after the modulation of SIRT3 levels in HCC cell lines (overexpression in Huh7, knockdown in HepG2). We also employed PD0332991 to modulate the SIRT3 expression in HCC cell and conducted functional assays. Results SIRT3 expression was downregulated in high glycolytic and proliferative HCC cells of human patients, xenograft model and HCC cell lines. Moreover, SIRT3 expression was downregulated after sorafenib treatment, resulting in reduced drug sensitivity in HCC cell lines. To enhance the anti-tumor effect of sorafenib, we employed PD0332991 (CDK4/6-Rb inhibitor) based on the correlation between SIRT3 and phosphorylated retinoblastoma protein in HCC. Notably, combined treatment with sorafenib and PD0332991 showed an enhancement of the anti-tumor effect in HCC cells. Conclusions Our data suggest that the modulation of SIRT3 by CDK4/6 inhibition might be useful for HCC therapy together with sorafenib, which, unfortunately, has limited efficacy and whose use is often associated with drug resistance.
Collapse
Affiliation(s)
- Hanhee Jo
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea.,Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 120-749, South Korea
| | - Yusun Park
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Taehun Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, South Korea
| | - Jisu Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Jong Sook Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Seon Yoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 120-749, South Korea
| | - Jee-In Chung
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 120-749, South Korea
| | - Hae Yong Ko
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 120-749, South Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, Seoul, South Korea
| | - Kyung Sik Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea.
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 120-749, South Korea.
| |
Collapse
|
99
|
Wu J, Deng Z, Sun M, Zhang W, Yang Y, Zeng Z, Wu J, Zhang Q, Liu Y, Chen Z, Guo X, Zhao KS, Huang Q, Chen Z. Polydatin protects against lipopolysaccharide-induced endothelial barrier disruption via SIRT3 activation. J Transl Med 2020; 100:643-656. [PMID: 31641228 DOI: 10.1038/s41374-019-0332-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/30/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
In a previous study, we demonstrated the role of polydatin (PD) in protecting against multiple organ dysfunction in sepsis. The aim of this study is to investigate whether PD protects against lipopolysaccharide (LPS)-induced endothelial barrier disruption through SIRT3 activation and to disclose the underlying mechanisms. Wild-type mice were injected with LPS and Evans Blue assay was performed to evaluate vascular permeability. Primary human umbilical vein endothelial cells (HUVECs) were stimulated with LPS. Endothelial permeability was evaluated by transendothelial electrical resistance (TER) and FITC-dextran leakage. SIRT3 activity was determined by a Deacetylase Fluorometric kit, and protein expression level of SIRT3 was detected by western blotting. Mitochondrial function was evaluated by determination of ROS level, mitochondrial membrane potential and mPTP opening. In endotoxemic mice, PD pretreatment attenuated vascular leakage in multiple organs while SIRT3 inhibition with 3-TYP reversed the effects of PD. PD treatment in late sepsis also exhibited barrier protective effects. In HUVECs, PD alleviated LPS-induced F-actin rearrangement, cadherin-catenin complex dissociation and endothelial hyperpermeability, whereas 3-TYP or SIRT3 siRNA attenuated the protective effects of PD. PD enhanced SIRT3 deacetylase activity, and attenuated LPS-induced decrease in SIRT3 expression as well. Furthermore, gain-of-function and loss-of-function strategies also confirmed the role of SIRT3 in enhancing endothelial barrier integrity. It was further ascertained that PD enhanced SIRT3-mediated deacetylation of SOD2 and cyclophilin D (CypD), thus suppressing mitochondrial dysfunction and subsequent endothelial barrier dysfunction. In addition, it was revealed that RAGE was involved in LPS-regulated SIRT3 signaling. Our results suggest that polydatin protects against LPS-induced endothelial barrier disruption dependent on SIRT3 and can be applied as a potential therapy for sepsis.
Collapse
Affiliation(s)
- Jie Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Zhiya Deng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Maomao Sun
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Weijin Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China.,Department of Geriatrics, Shantou Central Hospital, Shantou, 515000, China
| | - Yang Yang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Critical Care Medicine, the First People's Hospital of Chenzhou, Chenzhou, 423000, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Jianhua Wu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qin Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenfeng Chen
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Xiaohua Guo
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Ke-Seng Zhao
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China.
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
100
|
Xu J, Kitada M, Koya D. The impact of mitochondrial quality control by Sirtuins on the treatment of type 2 diabetes and diabetic kidney disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165756. [PMID: 32147421 DOI: 10.1016/j.bbadis.2020.165756] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/23/2020] [Accepted: 02/28/2020] [Indexed: 12/20/2022]
Abstract
The incidence of type 2 diabetes mellitus (T2DM) and diabetic kidney disease (DKD) has significantly increased worldwide in recent decades, and improved treatments for T2DM and DKD are urgently needed. The pathogenesis of aging-related disorders, such as T2DM and DKD, involves multiple mechanisms, including inflammation, autophagy impairment, and oxidative stress, which are closely associated with mitochondrial dysfunction. Therefore, mitochondrial quality control may be a novel therapeutic target for T2DM and DKD. Previous reports have shown that members of the mammalian Sirtuin family, SIRT 1-7, which are recognized as antiaging molecules, play a crucial role in the regulation of mitochondrial function and quality control through the modulation of oxidative stress, inflammation and autophagy. In this review, we summarized the research published in recent years to highlight the role of Sirtuins in mitochondrial quality control as a therapeutic target for T2DM and DKD.
Collapse
Affiliation(s)
- Jing Xu
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan; Department of Endocrinology and Metabolism, The Affiliated Hospital of Guizhou Medical University, NO. 28, Guiyi Street, Guiyang, Guizhou 550004, China
| | - Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan; Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan.
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan; Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|