1
|
D'Agnano V, Mariniello DF, Pagliaro R, Far MS, Schiattarella A, Scialò F, Stella G, Matera MG, Cazzola M, Bianco A, Perrotta F. Sirtuins and Cellular Senescence in Patients with Idiopathic Pulmonary Fibrosis and Systemic Autoimmune Disorders. Drugs 2024; 84:491-501. [PMID: 38630364 PMCID: PMC11189987 DOI: 10.1007/s40265-024-02021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 06/22/2024]
Abstract
The sirtuin family is a heterogeneous group of proteins that play a critical role in many cellular activities. Several degenerative diseases have recently been linked to aberrant sirtuin expression and activity because of the involvement of sirtuins in maintaining cell longevity and their putative antiaging function. Idiopathic pulmonary fibrosis and progressive pulmonary fibrosis associated with systemic autoimmune disorders are severe diseases characterized by premature and accelerated exhaustion and failure of alveolar type II cells combined with aberrant activation of fibroblast proliferative pathways leading to dramatic destruction of lung architecture. The mechanisms underlying alveolar type II cell exhaustion in these disorders are not fully understood. In this review, we have focused on the role of sirtuins in the pathogenesis of idiopathic and secondary pulmonary fibrosis and their potential as biomarkers in the diagnosis and management of fibrotic interstitial lung diseases.
Collapse
Affiliation(s)
- Vito D'Agnano
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
- U.O.C. Clinica Pneumologica L. Vanvitelli, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Domenica Francesca Mariniello
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
- U.O.C. Clinica Pneumologica L. Vanvitelli, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Raffaella Pagliaro
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
- U.O.C. Clinica Pneumologica L. Vanvitelli, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Mehrdad Savabi Far
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
| | - Angela Schiattarella
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
- U.O.C. Clinica Pneumologica L. Vanvitelli, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Filippo Scialò
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
| | - Giulia Stella
- Unit of Respiratory System Diseases, Department of Medical Sciences and Infectious Diseases, Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'L. Vanvitelli', Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
- U.O.C. Clinica Pneumologica L. Vanvitelli, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania 'L. Vanvitelli', Naples, Italy
- U.O.C. Clinica Pneumologica L. Vanvitelli, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| |
Collapse
|
2
|
Bursch KL, Goetz CJ, Smith BC. Current Trends in Sirtuin Activator and Inhibitor Development. Molecules 2024; 29:1185. [PMID: 38474697 DOI: 10.3390/molecules29051185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Sirtuins are NAD+-dependent protein deacylases and key metabolic regulators, coupling the cellular energy state with selective lysine deacylation to regulate many downstream cellular processes. Humans encode seven sirtuin isoforms (Sirt1-7) with diverse subcellular localization and deacylase targets. Sirtuins are considered protective anti-aging proteins since increased sirtuin activity is canonically associated with lifespan extension and decreased activity with developing aging-related diseases. However, sirtuins can also assume detrimental cellular roles where increased activity contributes to pathophysiology. Modulation of sirtuin activity by activators and inhibitors thus holds substantial potential for defining the cellular roles of sirtuins in health and disease and developing therapeutics. Instead of being comprehensive, this review discusses the well-characterized sirtuin activators and inhibitors available to date, particularly those with demonstrated selectivity, potency, and cellular activity. This review also provides recommendations regarding the best-in-class sirtuin activators and inhibitors for practical research as sirtuin modulator discovery and refinement evolve.
Collapse
Affiliation(s)
- Karina L Bursch
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Structural Genomics Unit, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Christopher J Goetz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Structural Genomics Unit, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
3
|
Gladwell LR, Ahiarah C, Rasheed S, Rahman SM, Choudhury M. Traditional Therapeutics and Potential Epidrugs for CVD: Why Not Both? Life (Basel) 2023; 14:23. [PMID: 38255639 PMCID: PMC10820772 DOI: 10.3390/life14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. In addition to the high mortality rate, people suffering from CVD often endure difficulties with physical activities and productivity that significantly affect their quality of life. The high prevalence of debilitating risk factors such as obesity, type 2 diabetes mellitus, smoking, hypertension, and hyperlipidemia only predicts a bleak future. Current traditional CVD interventions offer temporary respite; however, they compound the severe economic strain of health-related expenditures. Furthermore, these therapeutics can be prescribed indefinitely. Recent advances in the field of epigenetics have generated new treatment options by confronting CVD at an epigenetic level. This involves modulating gene expression by altering the organization of our genome rather than altering the DNA sequence itself. Epigenetic changes are heritable, reversible, and influenced by environmental factors such as medications. As CVD is physiologically and pathologically diverse in nature, epigenetic interventions can offer a ray of hope to replace or be combined with traditional therapeutics to provide the prospect of addressing more than just the symptoms of CVD. This review discusses various risk factors contributing to CVD, perspectives of current traditional medications in practice, and a focus on potential epigenetic therapeutics to be used as alternatives.
Collapse
Affiliation(s)
- Lauren Rae Gladwell
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Chidinma Ahiarah
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shireen Rasheed
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| |
Collapse
|
4
|
Wang LF, Li Q, Wen K, Zhao QH, Zhang YT, Zhao JL, Ding Q, Guan XH, Xiao YF, Deng KY, Xin HB. CD38 Deficiency Alleviates Diabetic Cardiomyopathy by Coordinately Inhibiting Pyroptosis and Apoptosis. Int J Mol Sci 2023; 24:16008. [PMID: 37958991 PMCID: PMC10650707 DOI: 10.3390/ijms242116008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetic cardiomyopathy is one of the diabetes mellitus-induced cardiovascular complications that can result in heart failure in severe cases, which is characterized by cardiomyocyte apoptosis, local inflammation, oxidative stress, and myocardial fibrosis. CD38, a main hydrolase of NAD+ in mammals, plays an important role in various cardiovascular diseases, according to our previous studies. However, the role of CD38 in diabetes-induced cardiomyopathy is still unknown. Here, we report that global deletion of the CD38 gene significantly prevented diabetic cardiomyopathy induced by high-fat diet plus streptozotocin (STZ) injection in CD38 knockout (CD38-KO) mice. We observed that CD38 expression was up-regulated, whereas the expression of Sirt3 was down-regulated in the hearts of diabetic mice. CD38 deficiency significantly promoted glucose metabolism and improved cardiac functions, exemplified by increased left ventricular ejection fraction and fractional shortening. In addition, we observed that CD38 deficiency markedly decreased diabetes or high glucose and palmitic acid (HG + PA)-induced pyroptosis and apoptosis in CD38 knockout hearts or cardiomyocytes, respectively. Furthermore, we found that the expression levels of Sirt3, mainly located in mitochondria, and its target gene FOXO3a were increased in CD38-deficient hearts and cardiomyocytes with CD38 knockdown under diabetic induction conditions. In conclusion, we demonstrated that CD38 deficiency protected mice from diabetes-induced diabetic cardiomyopathy by reducing pyroptosis and apoptosis via activating NAD+/Sirt3/FOXO3a signaling pathways.
Collapse
Affiliation(s)
- Ling-Fang Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.L.); (K.W.); (Q.-H.Z.); (Y.-T.Z.); (J.-L.Z.); (Q.D.); (X.-H.G.); (Y.-F.X.); (K.-Y.D.)
| | | | | | | | | | | | | | | | | | | | - Hong-Bo Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.L.); (K.W.); (Q.-H.Z.); (Y.-T.Z.); (J.-L.Z.); (Q.D.); (X.-H.G.); (Y.-F.X.); (K.-Y.D.)
| |
Collapse
|
5
|
Sosa Ponce ML, Remedios MH, Moradi-Fard S, Cobb JA, Zaremberg V. SIR telomere silencing depends on nuclear envelope lipids and modulates sensitivity to a lysolipid. J Cell Biol 2023; 222:e202206061. [PMID: 37042812 PMCID: PMC10103788 DOI: 10.1083/jcb.202206061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/29/2022] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
The nuclear envelope (NE) is important in maintaining genome organization. The role of lipids in communication between the NE and telomere regulation was investigated, including how changes in lipid composition impact gene expression and overall nuclear architecture. Yeast was treated with the non-metabolizable lysophosphatidylcholine analog edelfosine, known to accumulate at the perinuclear ER. Edelfosine induced NE deformation and disrupted telomere clustering but not anchoring. Additionally, the association of Sir4 at telomeres decreased. RNA-seq analysis showed altered expression of Sir-dependent genes located at sub-telomeric (0-10 kb) regions, consistent with Sir4 dispersion. Transcriptomic analysis revealed that two lipid metabolic circuits were activated in response to edelfosine, one mediated by the membrane sensing transcription factors, Spt23/Mga2, and the other by a transcriptional repressor, Opi1. Activation of these transcriptional programs resulted in higher levels of unsaturated fatty acids and the formation of nuclear lipid droplets. Interestingly, cells lacking Sir proteins displayed resistance to unsaturated-fatty acids and edelfosine, and this phenotype was connected to Rap1.
Collapse
Affiliation(s)
| | | | - Sarah Moradi-Fard
- Departments of Biochemistry and Molecular Biology and Oncology, Cumming School of Medicine, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Calgary, Canada
| | - Jennifer A. Cobb
- Departments of Biochemistry and Molecular Biology and Oncology, Cumming School of Medicine, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Calgary, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| |
Collapse
|
6
|
Chen Y, Huang Q, Feng Y. Exercise improves cardiac function in the aged rats with myocardial infarction. Physiol Res 2023; 72:27-35. [PMID: 36545879 PMCID: PMC10069814 DOI: 10.33549/physiolres.934966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
Abstract
Exercise can improve the cardiovascular health. However, the mechanism contributing to its beneficial effect on elderly patients with myocardial infarction is obscure. 20-month-old male Sprague-Dawley rats were used to establish myocardial infarction (MI) model by permanent ligation of the left anterior descending coronary artery (LAD) of the heart, followed by 4-week interval exercise training on a motor-driven rodent treadmill. The cardiac function, myocardial fibrosis, apoptosis, oxidative stress, and inflammatory responses were determined by using pressure transducer catheter, polygraph physiological data acquisition system, Masson's trichrome staining, and ELISA to evaluate the impact of post-MI exercise training on MI. Western blot were performed to detect the activation of AMPK/SIRT1/PGC-1alpha signaling in the hearts of aged rats. Exercise training significantly improved cardiac function and reduced the cardiac fibrosis. In infarcted heart, the apoptosis, oxidative stress, and inflammation were significantly reduced after 4-week exercise training. Mechanistically, AMPK/SIRT1/PGC-1alpha pathway was activated in the myocardial infarction area after exercise training, which might participate in the protection of cardiac function. Exercise training improves cardiac function in MI rats through reduction of apoptosis, oxidative stress, and inflammation, which may mediate by the activation of AMPK/SIRT1/PGC-1alpha signaling pathway.
Collapse
Affiliation(s)
- Y Chen
- Department of Geriatric Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China, Department of Cardiology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China.
| | | | | |
Collapse
|
7
|
Wang X, Huang Y, Zhang K, Chen F, Nie T, Zhao Y, He F, Ni J. Changes of energy metabolism in failing heart and its regulation by SIRT3. Heart Fail Rev 2023:10.1007/s10741-023-10295-5. [PMID: 36708431 DOI: 10.1007/s10741-023-10295-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 01/29/2023]
Abstract
Heart failure (HF) is the leading cause of hospitalization in elderly patients and a disease with extremely high morbidity and mortality rate worldwide. Although there are some existing treatment methods for heart failure, due to its complex pathogenesis and often accompanied by various comorbidities, there is still a lack of specific drugs to treat HF. The mortality rate of patients with HF is still high, highlighting an urgent need to elucidate the pathophysiological mechanisms of HF and seek new therapeutic approaches. The heart is an organ with a very high metabolic intensity, mainly using fatty acids, glucose, ketone bodies, and branched-chain amino acids as energy substrates to supply energy for the heart. Loss of metabolic flexibility and metabolic remodeling occurs with HF. Sirtuin3 (SIRT3) is a member of the NAD+-dependent Sirtuin family located in mitochondria, and can participate in mitochondrial physiological functions through the deacetylation of metabolic and respiratory enzymes in mitochondria. As the center of energy metabolism, mitochondria are involved in many physiological processes. Maintaining stable metabolic and physiological functions of the heart depends on normal mitochondrial function. The damage or loss of SIRT3 can lead to various cardiovascular diseases. Therefore, we summarize the recent progress of SIRT3 in cardiac mitochondrial protection and metabolic remodeling.
Collapse
Affiliation(s)
- Xiao Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yuting Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, 341000, China
| | - Kai Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Feng Chen
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Tong Nie
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yun Zhao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Feng He
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, 438000, China.
| | - Jingyu Ni
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
8
|
Doxorubicin induced cardio toxicity through sirtuins mediated mitochondrial disruption. Chem Biol Interact 2022; 365:110028. [DOI: 10.1016/j.cbi.2022.110028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 12/06/2022]
|
9
|
Li C, Jiang S, Wang H, Wang Y, Han Y, Jiang J. Berberine exerts protective effects on cardiac senescence by regulating the Klotho/SIRT1 signaling pathway. Biomed Pharmacother 2022; 151:113097. [PMID: 35609366 DOI: 10.1016/j.biopha.2022.113097] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 11/15/2022] Open
Abstract
Berberine (BBR), an isoquinoline alkaloid, exerts protective effects on various cardiac injuries, and also extends the lifespan of individuals. However, the cardioprotective effect of BBR on cardiac senescence remains unknown. This study investigated the effects of BBR on cardiac senescence and its underlying mechanism. Senescent H9c2 cells induced by doxorubicin (DOX) and naturally aged rats were used to evaluate the protective effects of BBR on cardiac senescence. The results showed that BBR protected H9c2 cells against DOX-induced senescence. Exogenous Klotho (KL) exerts similar effects to those of BBR. BBR significantly increased in protein expression of KL, while transfection with KL-specific siRNA (siKL) inhibited the protective effect of BBR against senescence. Both BBR and exogenous KL decreased the levels of reactive oxygen species, inhibited apoptosis, and alleviated mitochondrial dysfunction in these cells; and transfection with siKL attenuated these effects of BBR. In naturally aged rats, BBR indeed protected the animals from cardiac aging, at least partially, through lowering the levels of cardiac hypertrophy markers, and increased the expression of KL in cardiac tissue. Additionally, BBR markedly reversed downregulation of sirtuin1 (SIRTI) in the aged heart. In vitro experiments revealed that BBR and exogenous KL also increased the expression of SIRT1, whereas siKL limited this effect of BBR in senescent H9c2 cell. In summary, BBR upregulated KL expression and prevented heart from cardiac senescence through anti-oxidative and anti-apoptotic effects, as well as alleviation of mitochondrial dysfunction. These effects may be mediated via regulation of the Klotho/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Cong Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China
| | - Shuang Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China
| | - Hengfei Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China.
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China.
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China.
| |
Collapse
|
10
|
Charążka B, Siejka A. Correlations between serum sirtuin levels and cardiovascular risk factors in women with polycystic ovary syndrome. Adv Med Sci 2022; 67:123-128. [PMID: 35134601 DOI: 10.1016/j.advms.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/23/2021] [Accepted: 01/28/2022] [Indexed: 11/15/2022]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is associated with an increased risk of cardiovascular disease (CVD) and metabolic complications often related to obesity, such as insulin resistance and type 2 diabetes mellitus (T2DM). Sirtuins (SIRTs) are nicotinamide adenine dinucleotide NAD+-dependent histone deacetylases, which modulate protein post-translational modifications (PTMs), gene transcription, increase repairing activities of DNA, and regulate metabolic processes. The aim of our study was to measure the serum levels of SIRTs 1, 2, 3, 6 and 7, and evaluate correlations between SIRTs levels and cardiovascular risk factors in women with PCOS. PATIENTS AND METHODS The study included 54 women with PCOS and 33 healthy volunteers. Concentrations of SIRTs were measured by ELISA technique. RESULTS Mean serum levels of SIRTs did not differ significantly between PCOS and controls. SIRTs 1, 2, and 6 positively correlated with HDL, while SIRTs 2 and 6 negatively correlated with LDL cholesterol. Negative correlation between SIRT 3 and HOMA-IR and negative correlations of SIRT 1 and 2 with insulin 120' in oral glucose tolerance test (OGTT) were demonstrated. A positive correlation between BMI and SIRT 7 concentration, and a positive correlation between body weight and SIRT 7 concentration were verified in the study group. The observed correlations between concentrations of SIRTs and metabolic parameters may indicate the involvement of these factors in the development of cardiometabolic complications. CONCLUSIONS The results may indicate the involvement of SIRTs in the development of cardiometabolic complications. However, additional studies are required to validate this observation.
Collapse
Affiliation(s)
- Beata Charążka
- Endocrinology Outpatient Clinic, Central Teaching Hospital, Lodz, Poland
| | - Agnieszka Siejka
- Department of Clinical Endocrinology, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
11
|
Cui YK, Hong YX, Wu WY, Han WM, Wu Y, Wu C, Li GR, Wang Y. Acacetin ameliorates cardiac hypertrophy by activating Sirt1/AMPK/PGC-1α pathway. Eur J Pharmacol 2022; 920:174858. [PMID: 35219729 DOI: 10.1016/j.ejphar.2022.174858] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 12/20/2022]
Abstract
Cardiac hypertrophy is a major risk factor for developing heart failure. This study investigates the effects of the natural flavone acacetin on myocardial hypertrophy in cellular level and whole animals. In cardiomyocytes from neonatal rat with hypertrophy induced by angiotensin II (Ang II), acacetin at 0.3, 1, and 3 μM reduced the increased myocyte surface area, brain natriuretic peptide (BNP), and ROS production by upregulating anti-oxidative molecules (i.e. Nrf2, SOD1, SOD2, HO-1), anti-apoptotic protein Bcl-2, and downregulating the pro-apoptotic protein Bax and the inflammatory cytokine IL-6 in a concentration-dependent manner. In addition, acacetin rescued Ang II-induced impairment of PGC-1α, PPARα and pAMPK. These beneficial effects of acacetin were mediated by activation of Sirt1, which was confirmed in cardiac hypertrophy induced by abdominal aorta constriction (AAC) in SD rats. Acacetin prodrug (10 mg/kg, s.c., b.i.d.) treatment reduced the elevated artery blood pressure, improved the increased heart size and thickness of left ventricular wall and the ventricular fibrosis associated with inhibiting myocardial fibrosis and BNP, and reversed the impaired protective signal molecules including PGC-1α, Nrf2, PPARα, pAMPK and Sirt1 of left ventricular tissue. Our results demonstrate the novel pharmacological effect that acacetin ameliorates cardiac hypertrophy via Sirt1-mediated activation of AMPK/PGC-1α signal molecules followed by reducing oxidation, inflammation and apoptosis.
Collapse
Affiliation(s)
- Yu-Kai Cui
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Wei-Min Han
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Yao Wu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China; Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu, 210032, China.
| | - Yan Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China.
| |
Collapse
|
12
|
Sharma R, Sharma S, Thakur A, Singh A, Singh J, Nepali K, Liou JP. The Role of Epigenetic Mechanisms in Autoimmune, Neurodegenerative, Cardiovascular, and Imprinting Disorders. Mini Rev Med Chem 2022; 22:1977-2011. [PMID: 35176978 DOI: 10.2174/1389557522666220217103441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/01/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
Epigenetic mutations like aberrant DNA methylation, histone modifications, or RNA silencing are found in a number of human diseases. This review article discusses the epigenetic mechanisms involved in neurodegenerative disorders, cardiovascular disorders, auto-immune disorder, and genomic imprinting disorders. In addition, emerging epigenetic therapeutic strategies for the treatment of such disorders are presented. Medicinal chemistry campaigns highlighting the efforts of the chemists invested towards the rational design of small molecule inhibitors have also been included. Pleasingly, several classes of epigenetic inhibitors, DNMT, HDAC, BET, HAT, and HMT inhibitors along with RNA based therapies have exhibited the potential to emerge as therapeutics in the longer run. It is quite hopeful that epigenetic modulator-based therapies will advance to clinical stage investigations by leaps and bounds.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Arshdeep Singh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jagjeet Singh
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia.,Department of Pharmacy, Rayat-Bahara Group of Institutes, Hoshiarpur, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
da Silva JS, Gonçalves RGJ, Vasques JF, Rocha BS, Nascimento-Carlos B, Montagnoli TL, Mendez-Otero R, de Sá MPL, Zapata-Sudo G. Mesenchymal Stem Cell Therapy in Diabetic Cardiomyopathy. Cells 2022; 11:cells11020240. [PMID: 35053356 PMCID: PMC8773977 DOI: 10.3390/cells11020240] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
The incidence and prevalence of diabetes mellitus (DM) are increasing worldwide, and the resulting cardiac complications are the leading cause of death. Among these complications is diabetes-induced cardiomyopathy (DCM), which is the consequence of a pro-inflammatory condition, oxidative stress and fibrosis caused by hyperglycemia. Cardiac remodeling will lead to an imbalance in cell survival and death, which can promote cardiac dysfunction. Since the conventional treatment of DM generally does not address the prevention of cardiac remodeling, it is important to develop new alternatives for the treatment of cardiovascular complications induced by DM. Thus, therapy with mesenchymal stem cells has been shown to be a promising approach for the prevention of DCM because of their anti-apoptotic, anti-fibrotic and anti-inflammatory effects, which could improve cardiac function in patients with DM.
Collapse
Affiliation(s)
- Jaqueline S. da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
| | - Renata G. J. Gonçalves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-170, RJ, Brazil; (R.G.J.G.); (R.M.-O.)
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-170, RJ, Brazil;
| | - Bruna S. Rocha
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
| | - Bianca Nascimento-Carlos
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
| | - Tadeu L. Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
| | - Rosália Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-170, RJ, Brazil; (R.G.J.G.); (R.M.-O.)
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil
| | - Mauro P. L. de Sá
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
- Correspondence: or ; Tel.: +55-21-39386505
| |
Collapse
|
14
|
Meng F, Zhou J, Cheng X, Xu J, Kang L, Li D, Wang D, Bi Y. Design, Synthesis and Cardioprotection of Cryptotanshinone Derivatives. CHINESE J ORG CHEM 2022. [DOI: 10.6023/cjoc202110004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
15
|
Abdelnabi ALSM, Esmayel IM, Hussein S, Ali RM, AbdelAal AA. Sirtuin-1 in Egyptian patients with coronary artery disease. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021. [DOI: 10.1186/s43088-021-00164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Coronary artery disease (CAD) represents the leading cause of death worldwide. Animal and human studies have demonstrated that silent information regulator 1 (SIRT1) is involved in a wide range of physiological and pathological processes. This study aimed to measure the plasma level of SIRT1 in patients with CAD and explore its correlation with cardiovascular risk factors.
Results
Plasma SIRT1 was significantly lower in patients with chronic coronary syndrome (CCS) than in those in the control group and was significantly lower in patients with both acute myocardial infarction and unstable angina than in those in the control group and with CCS. Moreover, plasma SIRT1 was positively correlated with platelet count and negatively correlated with cholesterol and triglyceride levels.
Conclusions
The plasma level of SIRT1 is lower in patients with CAD compared to control and it could be a possible marker for this disease. Multi-center studies with follow-up measurements are recommended for further investigation.
Collapse
|
16
|
Gupta R, Ambasta RK, Kumar P. Multifaced role of protein deacetylase sirtuins in neurodegenerative disease. Neurosci Biobehav Rev 2021; 132:976-997. [PMID: 34742724 DOI: 10.1016/j.neubiorev.2021.10.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 01/07/2023]
Abstract
Sirtuins, a class III histone/protein deacetylase, is a central regulator of metabolic function and cellular stress response. This plays a pivotal role in the pathogenesis and progression of diseases such as cancer, neurodegeneration, metabolic syndromes, and cardiovascular disease. Sirtuins regulate biological and cellular processes, for instance, mitochondrial biogenesis, lipid and fatty acid oxidation, oxidative stress, gene transcriptional activity, apoptosis, inflammatory response, DNA repair mechanism, and autophagic cell degradation, which are known components for the progression of the neurodegenerative diseases (NDDs). Emerging evidence suggests that sirtuins are the useful molecular targets against NDDs like, Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD), and Amyotrophic Lateral Sclerosis (ALS). However, the exact mechanism of neuroprotection mediated through sirtuins remains unsettled. The manipulation of sirtuins activity with its modulators, calorie restriction (CR), and micro RNAs (miR) is a novel therapeutic approach for the treatment of NDDs. Herein, we reviewed the current putative therapeutic role of sirtuins in regulating synaptic plasticity and cognitive functions, which are mediated through the different molecular phenomenon to prevent neurodegeneration. We also explained the implications of sirtuin modulators, and miR based therapies for the treatment of life-threatening NDDs.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
17
|
Zhu HZ, Zhang LY, Zhai ME, Xia L, Cao Y, Xu L, Li KF, Jiang LQ, Shi H, Li X, Zhou YN, Ding W, Wang DX, Gao EH, Liu JC, Yu SQ, Duan WX. GDF11 Alleviates Pathological Myocardial Remodeling in Diabetic Cardiomyopathy Through SIRT1-Dependent Regulation of Oxidative Stress and Apoptosis. Front Cell Dev Biol 2021; 9:686848. [PMID: 34262905 PMCID: PMC8273395 DOI: 10.3389/fcell.2021.686848] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor β superfamily that alleviates cardiac hypertrophy, myocardial infarction, and vascular injury by regulating oxidative stress, inflammation, and cell survival. However, the roles and underlying mechanisms of GDF11 in diabetic cardiomyopathy (DCM) remain largely unknown. In this study, we sought to determine whether GDF11 could prevent DCM. After establishing a mouse model of diabetes by administering a high-fat diet and streptozotocin, intramyocardial injection of an adeno-associated virus was used to achieve myocardium-specific GDF11 overexpression. GDF11 remarkably improved cardiac dysfunction and interstitial fibrosis by reducing the levels of reactive oxygen species and protecting against cardiomyocyte loss. Mechanistically, decreased sirtuin 1 (SIRT1) expression and activity were observed in diabetic mice, which was significantly increased after GDF11 overexpression. To further explore how SIRT1 mediates the role of GDF11, the selective inhibitor EX527 was used to block SIRT1 signaling pathway, which abolished the protective effects of GDF11 against DCM. In vitro studies confirmed that GDF11 protected against H9c2 cell injury in high glucose and palmitate by attenuating oxidative injury and apoptosis, and these effects were eliminated by SIRT1 depletion. Our results demonstrate for the first time that GDF11 protects against DCM by regulating SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Han-Zhao Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Li-Yun Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Meng-En Zhai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Lin Xia
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yu Cao
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, The Air Force Medical University, Xi'an, China
| | - Lu Xu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, The Air Force Medical University, Xi'an, China
| | - Kai-Feng Li
- Basic Medical Teaching Experiment Center, Basic Medical College, The Air Force Medical University, Xi'an, China
| | - Li-Qing Jiang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Heng Shi
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Xiang Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Ye-Nong Zhou
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Wei Ding
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Dong-Xu Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Er-He Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, United States
| | - Jin-Cheng Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Shi-Qiang Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| | - Wei-Xun Duan
- Department of Cardiovascular Surgery, The First Affiliated Hospital, The Air Force Medical University, Xi'an, China
| |
Collapse
|
18
|
Caldas APS, Rocha DMUP, Bressan J, Hermsdorff HHM. Dietary fatty acids as nutritional modulators of sirtuins: a systematic review. Nutr Rev 2021; 79:235-246. [PMID: 32403131 DOI: 10.1093/nutrit/nuaa007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
CONTEXT The sirtuins (SIRT1 to SIRT7) constitute a family of highly conserved nicotinamide adenine dinucleotide-dependent proteins. When activated, sirtuins control essential cellular processes to maintain metabolic homeostasis, while lack of expression of sirtuins has been related to chronic disease. OBJECTIVE The aim of this systematic review is to analyze the role of fat consumption as a modulator of human sirtuins. DATA SOURCES This review was conducted according to PRISMA guidelines. Studies were identified by searches of the electronic databases PubMed/MEDLINE, Scopus, and Web of Science. STUDY SELECTION Randomized clinical trials assessing the effect of fatty acid consumption on sirtuin mRNA expression, sirtuin protein expression, or sirtuin protein activity were eligible for inclusion. DATA EXTRACTION Two authors screened and determined the quality of the studies; disagreements were resolved by the third author. All authors compared the compiled data. RESULTS Seven clinical studies with 3 different types of interventions involving healthy and nonhealthy participants were selected. Only SIRT1 and SIRT3 were evaluated. Overall, the evidence from clinical studies to date is insufficient to understand how lipid consumption modulates sirtuins in humans. The best-characterized mechanism highlights oleic acid as a natural activator of SIRT1. CONCLUSION These results draw attention to a new field of interest in nutrition science. The possible activation of sirtuins by dietary fat manipulation may represent an important nutritional strategy for management of chronic and metabolic disease. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration number CRD42018114456.
Collapse
Affiliation(s)
- Ana Paula S Caldas
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Daniela Mayumi U P Rocha
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | |
Collapse
|
19
|
Palomer X, Aguilar-Recarte D, García R, Nistal JF, Vázquez-Carrera M. Sirtuins: To Be or Not To Be in Diabetic Cardiomyopathy. Trends Mol Med 2021; 27:554-571. [PMID: 33839024 DOI: 10.1016/j.molmed.2021.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy is the leading cause of death among people with diabetes. Despite its severity and poor prognosis, there are currently no approved specific drugs to prevent or even treat diabetic cardiomyopathy. There is a need to understand the pathogenic mechanisms underlying the development of diabetic cardiomyopathy to design new therapeutic strategies. These mechanisms are complex and intricate and include metabolic dysregulation, inflammation, oxidative stress, fibrosis, and apoptosis. Sirtuins, a group of deacetylase enzymes, play an important role in all these processes and are, therefore, potential molecular targets for treating this disease. In this review, we discuss the role of sirtuins in the heart, focusing on their contribution to the pathogenesis of diabetic cardiomyopathy and how their modulation could be therapeutically useful.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - David Aguilar-Recarte
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Raquel García
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - J Francisco Nistal
- Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Departamento de Ciencias Médicas y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL); Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Santander, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
20
|
Jayakumari NR, Rajendran RS, Sivasailam A, Parambil ST, Reghuvaran AC, Sreelatha HV, Gopala S. Honokiol regulates mitochondrial substrate utilization and cellular fatty acid metabolism in diabetic mice heart. Eur J Pharmacol 2021; 896:173918. [PMID: 33529726 DOI: 10.1016/j.ejphar.2021.173918] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/13/2021] [Accepted: 01/26/2021] [Indexed: 01/06/2023]
Abstract
Type 2 diabetes mellitus is strongly associated with cardiac mitochondrial dysfunction, which is one of the main reasons for cardiovascular diseases. Among the mitochondrial metabolic changes, fatty acid metabolism is of great importance as cardiac tissues depend primarily on fatty acids. Honokiol, a constituent of Magnolia tree bark extract, is reported to strongly influence cardiac mitochondrial functions, via various mechanisms. The current study showed that honokiol decreased fatty acid-mediated complex I respiration and increased carbohydrate-mediated complex I and II respiration in diabetic C57BL/6 mice cardiac mitochondria. It was also found that honokiol treatment decreased expression of Cluster of Differentiation 36, AMP-activated kinases and nuclear transcription factors like, Peroxisome proliferator-activated receptor γ co-activator 1α/β and Peroxisome proliferator-activated receptor α, surrogating the evidence of decreased fatty acid-mediated complex I respiration. Honokiol treatment also reduced the levels of mitochondrial acetylated proteins, suggesting the possible action of honokiol via acetylation/deacetylation mechanism of regulation of protein functions in diabetic mitochondria. The antioxidant effect of honokiol is evidenced by the augmented expression of Manganese super oxide dismutase. In conclusion, honokiol imparts beneficial effect on diabetic cardiac mitochondria by decreasing the oxidant burden via regulating mitochondrial fatty acid respiration and expression of oxidant response factors.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Acetylation
- Animals
- Antioxidants/pharmacology
- Biphenyl Compounds/pharmacology
- CD36 Antigens/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Energy Metabolism/drug effects
- Fatty Acids/metabolism
- Lignans/pharmacology
- Male
- Mice, Inbred C57BL
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- NF-E2-Related Factor 2/metabolism
- Oxidative Stress/drug effects
- PPAR alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Streptozocin
- Superoxide Dismutase/metabolism
- Mice
Collapse
Affiliation(s)
- Nandini Ravikumar Jayakumari
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Raji Sasikala Rajendran
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Ashok Sivasailam
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Sulfath Thottungal Parambil
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Anand Chellappan Reghuvaran
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Harikrishnan Vijayakumar Sreelatha
- Division of Laboratory Animal Sciences, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
21
|
Soni SK, Basu P, Singaravel M, Sharma R, Pandi-Perumal SR, Cardinali DP, Reiter RJ. Sirtuins and the circadian clock interplay in cardioprotection: focus on sirtuin 1. Cell Mol Life Sci 2021; 78:2503-2515. [PMID: 33388853 PMCID: PMC11073088 DOI: 10.1007/s00018-020-03713-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/09/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Chronic disruption of circadian rhythms which include intricate molecular transcription-translation feedback loops of evolutionarily conserved clock genes has serious health consequences and negatively affects cardiovascular physiology. Sirtuins (SIRTs) are nuclear, cytoplasmic and mitochondrial histone deacetylases that influence the circadian clock with clock-controlled oscillatory protein, NAMPT, and its metabolite NAD+. Sirtuins are linked to the multi-organ protective role of melatonin, particularly in acute kidney injury and in cardiovascular diseases, where melatonin, via upregulation of SIRT1 expression, inhibits the apoptotic pathway. This review focuses on SIRT1, an NAD+-dependent class III histone deacetylase which counterbalances the intrinsic histone acetyltransferase activity of one of the clock genes, CLOCK. SIRT1 is involved in the development of cardiomyocytes, regulation of voltage-gated cardiac sodium ion channels via deacetylation, prevention of atherosclerotic plaque formation in the cardiovascular system, protection against oxidative damage and anti-thrombotic actions. Overall, SIRT1 has a see-saw effect on cardioprotection, with low levels being cardioprotective and higher levels leading to cardiac hypertrophy.
Collapse
Affiliation(s)
- Sanjeev Kumar Soni
- Chronobiology Laboratory, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Priyoneel Basu
- Chronobiology Laboratory, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Muniyandi Singaravel
- Chronobiology Laboratory, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | | | - Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
22
|
Berberine-induced TFEB deacetylation by SIRT1 promotes autophagy in peritoneal macrophages. Aging (Albany NY) 2021; 13:7096-7119. [PMID: 33639613 PMCID: PMC7993719 DOI: 10.18632/aging.202566] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease that commonly affects the elderly and is characterized by vascular damage, macrophage infiltration, and plaque formation. Moreover, it increases the risk of cardiovascular disease. The pathogenesis of atherosclerosis involves an interplay between macrophage autophagy and apoptosis. A recently discovered transcription factor, transcription factor EB (TFEB) is known to activate autophagy in macrophages. Sirtuin deacetylase 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, activates several transcription factors, including TFEB. We studied the effects of berberine on the NAD+ synthesis pathway and interactions between SIRT1 and TFEB. We also studied the effects of berberine-induced TFEB activation via SIRT1 on autophagy and apoptosis of peritoneal macrophages. We found that berberine promoted autophagy of peritoneal macrophages by activating SIRT1 via the NAD+ synthesis pathway and, in turn, promoting TFEB nuclear translocation and deacetylation. The functional regulation of SIRT1 and TFEB by berberine could be exploited as a potential therapeutic strategy for atherosclerosis.
Collapse
|
23
|
Darvishzadeh Mahani F, Khaksari M, Raji-Amirhasani A. Renoprotective effects of estrogen on acute kidney injury: the role of SIRT1. Int Urol Nephrol 2021; 53:2299-2310. [PMID: 33458788 DOI: 10.1007/s11255-020-02761-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/15/2020] [Indexed: 01/23/2023]
Abstract
Acute kidney injury (AKI) is a common syndrome associated with high morbidity and mortality, despite progress in medical care. Many studies have shown that there are sex differences and different role of sex hormones particularly estrogens in kidney injury. In this regard, the incidence and rate of progression of kidney diseases are higher in men compared with women. These observations suggest that female sex hormone may be renoprotective. Silent information regulator 2 homolog 1 (SIRT1) is a histone deacetylase, which is implicated in multiple biologic processes in several organisms. In the kidneys, SIRT1 inhibits renal cell apoptosis, inflammation, and fibrosis. Studies have reported a link between SIRT1 and estrogen. In addition, SIRT1 regulates ERα expression and inhibition of SIRT1 activity suppresses ERα expression. This effect leads to inhibition of estrogen-responsive gene expression. In this text, we review the role of SIRT1 in mediating the protective effects of estrogen in the onset and progression of AKI.
Collapse
Affiliation(s)
- Fatemeh Darvishzadeh Mahani
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alireza Raji-Amirhasani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
24
|
Raj S, Dsouza LA, Singh SP, Kanwal A. Sirt6 Deacetylase: A Potential Key Regulator in the Prevention of Obesity, Diabetes and Neurodegenerative Disease. Front Pharmacol 2020; 11:598326. [PMID: 33442387 PMCID: PMC7797778 DOI: 10.3389/fphar.2020.598326] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022] Open
Abstract
Sirtuins, NAD + dependent proteins belonging to class III histone deacetylases, are involved in regulating numerous cellular processes including cellular stress, insulin resistance, inflammation, mitochondrial biogenesis, chromatin silencing, cell cycle regulation, transcription, and apoptosis. Of the seven mammalian sirtuins present in humans, Sirt6 is an essential nuclear sirtuin. Until recently, Sirt6 was thought to regulate chromatin silencing, but new research indicates its role in aging, diabetes, cardiovascular disease, lipid metabolism, neurodegenerative diseases, and cancer. Various murine models demonstrate that Sirt6 activation is beneficial in alleviating many disease conditions and increasing lifespan, showing that Sirt6 is a critical therapeutic target in the treatment of various disease conditions in humans. Sirt6 also regulates the pathogenesis of multiple diseases by acting on histone proteins and non-histone proteins. Endogenous and non-endogenous modulators regulate both activation and inhibition of Sirt6. Few Sirt6 specific non-endogenous modulators have been identified. Hence the identification of Sirt6 specific modulators may have potential therapeutic roles in the diseases described above. In this review, we describe the development of Sirt6, the role it plays in the human condition, the functional role and therapeutic importance in disease processes, and specific modulators and molecular mechanism of Sirt6 in the regulation of metabolic homeostasis, cardiovascular disease, aging, and neurodegenerative disease.
Collapse
Affiliation(s)
- Swapnil Raj
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Liston Augustine Dsouza
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Shailendra Pratap Singh
- Department of Biomedical Engineering, School of Engineering and Technology, Central University of Rajasthan, Kishangarh, India
| | - Abhinav Kanwal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| |
Collapse
|
25
|
Xu W, Li L, Zhang L. NAD + Metabolism as an Emerging Therapeutic Target for Cardiovascular Diseases Associated With Sudden Cardiac Death. Front Physiol 2020; 11:901. [PMID: 32903597 PMCID: PMC7438569 DOI: 10.3389/fphys.2020.00901] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
In addition to its central role in mediating oxidation reduction in fuel metabolism and bioenergetics, nicotinamide adenine dinucleotide (NAD+) has emerged as a vital co-substrate for a number of proteins involved in diverse cellular processes, including sirtuins, poly(ADP-ribose) polymerases and cyclic ADP-ribose synthetases. The connection with aging and age-associated diseases has led to a new wave of research in the cardiovascular field. Here, we review the basics of NAD+ homeostasis, the molecular physiology and new advances in ischemic-reperfusion injury, heart failure, and arrhythmias, all of which are associated with increased risks for sudden cardiac death. Finally, we summarize the progress of NAD+-boosting therapy in human cardiovascular diseases and the challenges for future studies.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Le Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
26
|
Mazumder S, Barman M, Bandyopadhyay U, Bindu S. Sirtuins as endogenous regulators of lung fibrosis: A current perspective. Life Sci 2020; 258:118201. [PMID: 32781070 DOI: 10.1016/j.lfs.2020.118201] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/26/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023]
Abstract
Fibrotic lung diseases qualify among the most dreaded irreversible interstitial pulmonary complications with progressive yet largely unpredictable clinical course. Idiopathic pulmonary fibrosis (IPF) is the most challenging prototype characterized by unknown and complex molecular etiology, severe dearth of non-invasive therapeutic options and average lifespan of 2-5 years in patients post diagnosis. Lung fibrosis (LF) is a leading cause of death in the industrialized world with the propensity to contract, significantly increasing with age. Approximately 45% deaths in US are attributed to fibrotic diseases while around 7% respiratory disease-associated deaths, annually in UK, are actually attributed to IPF. Recent developments in the field of LF have unambiguously pointed towards the pivotal role of Sirtuins (SIRTs) in regulating disease progression, thereby qualifying as potential anti-fibrotic drug targets. These NAD+-dependent lysine deacetylases, deacylases and ADP-ribosyltransferases are evolutionarily conserved proteins, regulated by diverse metabolic/environmental factors and implicated in age-related degenerative and inflammatory disorders. While SIRT1, SIRT6 and SIRT7 are predominantly nuclear, SIRT3, SIRT4, SIRT5 are mainly mitochondrial and SIRT2 is majorly cytosolic with occasional nuclear translocation. SIRT1, SIRT3, SIRT6 and SIRT7 are documented as cytoprotective sirtuins implicated in cardiovascular, pulmonary and metabolic diseases including fibrosis; however functional roles of remaining sirtuins in pulmonary pathologies are yet elusive. Here, we provide a comprehensive recent update on the regulatory role of sirtuins on LF along with discussion on potential therapeutic modulation of endogenous Sirtuin expression through synthetic/plant-derived compounds which can help synthetic chemists and ethnopharmacologists to design new-generation cheap, non-toxic Sirtuin-based drugs against LF.
Collapse
Affiliation(s)
- Somnath Mazumder
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Mukta Barman
- Department of Zoology, Cooch Behar Panchanan Barma University, Vivekananda Street, Cooch Behar, West Bengal 736101, India
| | - Uday Bandyopadhyay
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India; Division of Molecular Medicine, Bose Institute, P-1/12, CIT Rd, Scheme VIIM, Kankurgachi, Kolkata, West Bengal 700054, India
| | - Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Vivekananda Street, Cooch Behar, West Bengal 736101, India.
| |
Collapse
|
27
|
Devi K, Singh N, Jaggi AS. Dual role of sirtuin 1 in inflammatory bowel disease. Immunopharmacol Immunotoxicol 2020; 42:385-391. [PMID: 32619377 DOI: 10.1080/08923973.2020.1790595] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Silent information regulator-1 (SIRT-1), is a member of the class III group of histone deacetylases and is collectively called sirtuins. There have been preclinical and clinical studies indicating the downregulation and decreased activity of sirtuin 1 in various inflammatory bowel disease models. Furthermore, the downregulation of sirtuin 1 is responsible for the sustained production of proinflammatory cytokines and the generation of oxidative stress in colitis. Hyperacetylation of NF-κB and HSF-1 (heat shock factor-1) in the absence of sirtuin1 is responsible for the induction of colitis. Accordingly, exogenous administration of sirtuin1 activators has been shown to attenuate the colitis in various inflammatory bowel disease models. On the other hand, the knockdown of sirtuin 1 gene or pharmacologic inhibition of sirtuin 1 has also been shown to be protective in the colitis. The deletion of the sirtuin1 gene may be helpful in the improvement of the disease condition of colitis through the maintenance of gastrointestinal immune homeostasis. The current review highlights the dual role of sirtuin 1 in the different experimental models of IBD along with possible mechanisms.
Collapse
Affiliation(s)
- Karam Devi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
28
|
Evans LW, Athukorala M, Martinez-Guryn K, Ferguson BS. The Role of Histone Acetylation and the Microbiome in Phytochemical Efficacy for Cardiovascular Diseases. Int J Mol Sci 2020; 21:E4006. [PMID: 32503339 PMCID: PMC7313062 DOI: 10.3390/ijms21114006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/12/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVD) are the main cause of death worldwide and create a substantial financial burden. Emerging studies have begun to focus on epigenetic targets and re-establishing healthy gut microbes as therapeutic options for the treatment and prevention of CVD. Phytochemicals, commonly found in fruits and vegetables, have been shown to exert a protective effect against CVD, though their mechanisms of action remain incompletely understood. Of interest, phytochemicals such as curcumin, resveratrol and epigallocatechin gallate (EGCG) have been shown to regulate both histone acetylation and microbiome re-composition. The purpose of this review is to highlight the microbiome-epigenome axis as a therapeutic target for food bioactives in the prevention and/or treatment of CVD. Specifically, we will discuss studies that highlight how the three phytochemicals above alter histone acetylation leading to global changes in gene expression and CVD protection. Then, we will expand upon these phytochemicals to discuss the impact of phytochemical-microbiome-histone acetylation interaction in CVD.
Collapse
Affiliation(s)
- Levi W. Evans
- Department of Nutrition, University of Nevada Reno, Reno, NV 89557, USA; (L.W.E.); (M.A.)
| | - Maheshi Athukorala
- Department of Nutrition, University of Nevada Reno, Reno, NV 89557, USA; (L.W.E.); (M.A.)
| | | | - Bradley S. Ferguson
- Department of Nutrition, University of Nevada Reno, Reno, NV 89557, USA; (L.W.E.); (M.A.)
- Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada Reno, Reno, NV 89557, USA
| |
Collapse
|
29
|
Ramachandra CJA, Chua J, Cong S, Kp MMJ, Shim W, Wu JC, Hausenloy DJ. Human-induced pluripotent stem cells for modelling metabolic perturbations and impaired bioenergetics underlying cardiomyopathies. Cardiovasc Res 2020; 117:694-711. [PMID: 32365198 DOI: 10.1093/cvr/cvaa125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/23/2020] [Accepted: 04/24/2020] [Indexed: 12/17/2022] Open
Abstract
Normal cardiac contractile and relaxation functions are critically dependent on a continuous energy supply. Accordingly, metabolic perturbations and impaired mitochondrial bioenergetics with subsequent disruption of ATP production underpin a wide variety of cardiac diseases, including diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, anthracycline cardiomyopathy, peripartum cardiomyopathy, and mitochondrial cardiomyopathies. Crucially, there are no specific treatments for preventing the onset or progression of these cardiomyopathies to heart failure, one of the leading causes of death and disability worldwide. Therefore, new treatments are needed to target the metabolic disturbances and impaired mitochondrial bioenergetics underlying these cardiomyopathies in order to improve health outcomes in these patients. However, investigation of the underlying mechanisms and the identification of novel therapeutic targets have been hampered by the lack of appropriate animal disease models. Furthermore, interspecies variation precludes the use of animal models for studying certain disorders, whereas patient-derived primary cell lines have limited lifespan and availability. Fortunately, the discovery of human-induced pluripotent stem cells has provided a promising tool for modelling cardiomyopathies via human heart tissue in a dish. In this review article, we highlight the use of patient-derived iPSCs for studying the pathogenesis underlying cardiomyopathies associated with metabolic perturbations and impaired mitochondrial bioenergetics, as the ability of iPSCs for self-renewal and differentiation makes them an ideal platform for investigating disease pathogenesis in a controlled in vitro environment. Continuing progress will help elucidate novel mechanistic pathways, and discover novel therapies for preventing the onset and progression of heart failure, thereby advancing a new era of personalized therapeutics for improving health outcomes in patients with cardiomyopathy.
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jasper Chua
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Faculty of Science, National University of Singapore, 6 Science Drive 2, Singapore 117546, Singapore
| | - Shuo Cong
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Xuhui District, Shanghai 200032, China
| | - Myu Mai Ja Kp
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of Technology, 10 Dover Drive, Singapore 138683, Singapore
| | - Joseph C Wu
- Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine, Stanford University, Stanford, CA 94305, USA.,Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.,Yong Loo Lin Medical School, National University of Singapore, 10 Medical Drive, Singapore 11759, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, Bloomsbury, London WC1E 6HX, UK.,Cardiovascular Research Centre, College of Medical and Health Sciences, Asia University, No. 500, Liufeng Road, Wufeng District, Taichung City 41354,Taiwan
| |
Collapse
|
30
|
Effects of doxorubicin on the heart: From molecular mechanisms to intervention strategies. Eur J Pharmacol 2019; 866:172818. [PMID: 31758940 DOI: 10.1016/j.ejphar.2019.172818] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022]
Abstract
Cancer remains a major public health problem worldwide and was responsible for 9.6 million deaths in 2018. Oncologic treatments such as doxorubicin (Dox) and trastuzumab (Trz) are chemotherapeutic drugs used to treat several types of cancer, including solid and non-solid malignancies. Although these drugs have a significant impact on the reduction in mortality of cancer patients, this treatment has an adverse effect on the cardiovascular system. The mechanisms associated with Dox-induced cardiotoxicity involve inflammation, oxidative stress, apoptosis, mitochondria impairment and dysregulation of autophagy. Unfortunately, Trz, an effective anti-cancer drug, can potentiate these adverse effects. Trz is a recombinant DNA-derived humanized monoclonal antibody against human epidermal growth factor receptor 2 (HER2). Despite its high anti-cancer efficacy, Trz also has a cardiotoxic effect. Unlike Dox, this adverse effect of Trz on the heart is mostly reversible. A strategy to prevent this undesirable effect is urgently needed. Currently, several pharmacological interventions have shown promising results that might effectively attenuate Dox- and Trz-induced cardiac dysfunction. In this review, reports from in vitro, in vivo and clinical studies pertinent to the underlying mechanisms involved in chemotherapy-induced cardiotoxicity, are comprehensively summarized and discussed. In addition, the potential pharmacological interventions to prevent these cardiotoxic effects are described.
Collapse
|
31
|
Bugyei-Twum A, Ford C, Civitarese R, Seegobin J, Advani SL, Desjardins JF, Kabir G, Zhang Y, Mitchell M, Switzer J, Thai K, Shen V, Abadeh A, Singh KK, Billia F, Advani A, Gilbert RE, Connelly KA. Sirtuin 1 activation attenuates cardiac fibrosis in a rodent pressure overload model by modifying Smad2/3 transactivation. Cardiovasc Res 2019; 114:1629-1641. [PMID: 29800064 DOI: 10.1093/cvr/cvy131] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/17/2018] [Indexed: 12/14/2022] Open
Abstract
Aims Transforming growth factor β1 (TGF-β1) is a prosclerotic cytokine involved in cardiac remodelling leading to heart failure (HF). Acetylation/de-acetylation of specific lysine residues in Smad2/3 has been shown to regulate TGF-β signalling by altering its transcriptional activity. Recently, the lysine de-acetylase sirtuin 1 (SIRT1) has been shown to have a cardioprotective effect; however, SIRT1 expression and activity are paradoxically reduced in HF. Herein, we investigate whether pharmacological activation of SIRT1 would induce cardioprotection in a pressure overload model and assess the impact of SIRT1 activation on TGF-β signalling and the fibrotic response. Methods and results Eight weeks old male C57BL/6 mice were randomized to undergo sham surgery or transverse aortic constriction (TAC) to induce pressure overload. Post-surgery, animals were further randomized to receive SRT1720 or vehicle treatment. Echocardiography, pressure-volume loops, and histological analysis revealed an impairment in cardiac function and deleterious left ventricular remodelling in TAC-operated animals that was improved with SRT1720 treatment. Genetic ablation and cell culture studies using a Smad-binding response element revealed SIRT1 to be a specific target of SRT1720 and identified Smad2/3 as a SIRT1 specific substrate. Conclusion Overall, our data demonstrate that Smad2/3 is a specific SIRT1 target and suggests that pharmacological activation of SIRT1 may be a novel therapeutic strategy to prevent/reverse HF via modifying Smad activity.
Collapse
Affiliation(s)
- Antoinette Bugyei-Twum
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Christopher Ford
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Robert Civitarese
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Jessica Seegobin
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Jean-Francois Desjardins
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Golam Kabir
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Yanling Zhang
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Melissa Mitchell
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Jennifer Switzer
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Kerri Thai
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Vanessa Shen
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Armin Abadeh
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Krishna K Singh
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Filio Billia
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Advani
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Richard E Gilbert
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Kim A Connelly
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Abstract
Aging is associated with a progressive decline in cardiovascular structure and function. Accumulating evidence links cardiovascular aging to epigenetic alterations encompassing a complex interplay of DNA methylation, histone posttranslational modifications, and dynamic nucleosome occupancy governed by numerous epigenetic factors. Advances in genomics technology have led to a profound understanding of chromatin reorganization in both cardiovascular aging and diseases. This review summarizes recent discoveries in epigenetic mechanisms involved in cardiovascular aging and diseases and discusses potential therapeutic strategies to retard cardiovascular aging and conquer related diseases through the rejuvenation of epigenetic signatures to a young state.
Collapse
Affiliation(s)
- Weiqi Zhang
- From the Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China (W.Z., G.-H.L.).,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics (W.Z., G.-H.L.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology (M.S.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology (J.Q.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| | - Guang-Hui Liu
- From the Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China (W.Z., G.-H.L.).,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics (W.Z., G.-H.L.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| |
Collapse
|
33
|
Jia D, Hou L, Lv Y, Xi L, Tian Z. Postinfarction exercise training alleviates cardiac dysfunction and adverse remodeling via mitochondrial biogenesis and SIRT1/PGC-1α/PI3K/Akt signaling. J Cell Physiol 2019; 234:23705-23718. [PMID: 31187505 DOI: 10.1002/jcp.28939] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Exercise training mitigates cardiac pathological remodeling and dysfunction caused by myocardial infarction (MI), but its underlying cellular and molecular mechanisms remain elusive. Our present study in an in vivo rat model of MI determined the impact of post-MI exercise training on myocardial fibrosis, mitochondrial biogenesis, antioxidant capacity, and ventricular function. Adult male rats were randomized into: (a) Sedentary control group; (b) 4-week treadmill exercise training group; (c) Sham surgery group; (d) MI group with permanent ligation of left anterior descending coronary artery and kept sedentary during post-MI period; and (e) post-MI 4-week exercise training group. Results indicated that exercise training significantly improved post-MI left ventricular function and reduced markers of cardiac fibrosis. Exercise training also significantly attenuated MI-induced mitochondrial damage and oxidative stress, which were associated with enhanced antioxidant enzyme expression and/or activity and total antioxidant capacity in the heart. Interestingly, the adaptive activation of the SIRT1/PGC-1α/PI3K/Akt signaling following MI was further enhanced by post-MI exercise training, which is likely responsible for exercise-induced cardioprotection and mitochondrial biogenesis. In conclusion, this study has provided novel evidence on the activation of SIRT1/PGC-1α/PI3K/Akt pathway, which may mediate exercise-induced cardioprotection through reduction of cardiac fibrosis and oxidative stress, as well as improvement of mitochondrial integrity and biogenesis in post-MI myocardium.
Collapse
Affiliation(s)
- Dandan Jia
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Lu Hou
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Yongzhi Lv
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Lei Xi
- Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
34
|
Forini F, Nicolini G, Pitto L, Iervasi G. Novel Insight Into the Epigenetic and Post-transcriptional Control of Cardiac Gene Expression by Thyroid Hormone. Front Endocrinol (Lausanne) 2019; 10:601. [PMID: 31555215 PMCID: PMC6727178 DOI: 10.3389/fendo.2019.00601] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Thyroid hormone (TH) signaling is critically involved in the regulation of cardiovascular physiology. Even mild reductions of myocardial TH levels, as occur in hypothyroidism or low T3 state conditions, are thought to play a role in the progression of cardiac disorders. Due to recent advances in molecular mechanisms underlying TH action, it is now accepted that TH-dependent modulation of gene expression is achieved at multiple transcriptional and post-transcriptional levels and involves the cooperation of many processes. Among them, the epigenetic remodeling of chromatin structure and the interplay with non-coding RNA have emerged as novel TH-dependent pathways that add further degrees of complexity and broaden the network of genes controlled by TH signaling. Increasing experimental and clinical findings indicate that aberrant function of these regulatory mechanisms promotes the evolution of cardiac disorders such as post-ischemic injury, pathological hypertrophy, and heart failure, which may be reversed by the correction of the underlying TH dyshomeostasis. To encourage the clinical implementation of a TH replacement strategy in cardiac disease, here we discuss the crucial effect of epigenetic modifications and control of non-coding RNA in TH-dependent regulation of biological processes relevant for cardiac disease evolution.
Collapse
|
35
|
Godugu C, Kumari P, Khurana A. Nanoyttria attenuates isoproterenol-induced cardiac injury. Nanomedicine (Lond) 2018; 13:2961-2980. [DOI: 10.2217/nnm-2018-0223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: The present study was designed to probe the cardioprotective effects of nanoyttria (NY). Materials & methods: NY was characterized using various techniques. Isoproterenol (ISO)-induced cardiotoxicity challenged mice were treated with NY for 28 days at two doses (0.4 and 4 mg/kg, intraperitoneally). Results: NY demonstrated free radical scavenging activity as shown by a 2,2-diphenyl-1-picrylhydrazyl assay. NY treatment showed alleviation of ISO-induced cardiotoxicity as evident from the reduction in biochemical parameters. The expression of proinflammatory cytokines (IL-1β, IL-6 and TNF-α) showed significant decrease upon NY treatment. Histopathology and ECG showed protection in histoarchitecture and rhythm of heart, respectively. Reduction in hydroxyproline and TGF-β1 expression indicated antifibrotic activity. Conclusion: We report for the first time that NY ameliorates ISO-induced cardiac remodeling.
Collapse
Affiliation(s)
- Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana State, PIN-500037, India
| | - Preeti Kumari
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana State, PIN-500037, India
| | - Amit Khurana
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana State, PIN-500037, India
| |
Collapse
|
36
|
Parodi-Rullán RM, Chapa-Dubocq XR, Javadov S. Acetylation of Mitochondrial Proteins in the Heart: The Role of SIRT3. Front Physiol 2018; 9:1094. [PMID: 30131726 PMCID: PMC6090200 DOI: 10.3389/fphys.2018.01094] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022] Open
Abstract
A growing number of studies have demonstrated the role of post-translational modifications of proteins, particularly acetylation, in human diseases including neurodegenerative and cardiovascular diseases, diabetes, cancer, and in aging. Acetylation of mitochondrial proteins has been shown to be involved in the pathogenesis of cardiac diseases such as myocardial infarction (ischemia-reperfusion) and heart failure. Indeed, over 60% of mitochondrial proteins contain acetylation sites, and most of these proteins are involved in mitochondrial bioenergetics. Mitochondrial non-enzymatic acetylation is enabled by acetyl-coenzyme A abundance and serves as the primary pathway of acetylation in mitochondria. Hence, regulation of enzymatic deacetylation becomes the most important mechanism to control acetylation/deacetylation of mitochondrial proteins. Acetylation/deacetylation of mitochondrial proteins has been regarded as a key regulator of mitochondrial metabolism and function. Proteins are deacetylated by NAD+-dependent deacetylases known as sirtuins (SIRTs). Among seven sirtuin isoforms, only SIRT3, SIRT4, and SIRT5 are localized in the mitochondria. SIRT3 is the main mitochondrial sirtuin which plays a key role in maintaining metabolic and redox balance in the mitochondria under physiological and pathological conditions. SIRT3 regulates the enzymatic activity of proteins involved in fatty acid oxidation, tricarboxylic acid cycle, electron transport chain, and oxidative phosphorylation. Although many enzymes have been identified as targets for SIRT3, cardiac-specific SIRT3 effects and regulations could differ from those in non-cardiac tissues. Therefore, it is important to elucidate the contribution of SIRT3 and mitochondrial protein acetylation/deacetylation in mitochondrial metabolism and cardiac dysfunction. Here, we summarize previous studies and provide a comprehensive analysis of the role of SIRT3 in mitochondria metabolism and bioenergetics under physiological conditions and in cardiac diseases. In addition, the review discusses mitochondrial protein acetylation as a potential target for cardioprotection.
Collapse
Affiliation(s)
- Rebecca M Parodi-Rullán
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, United States
| | - Xavier R Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, United States
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, United States
| |
Collapse
|
37
|
Zullo A, Simone E, Grimaldi M, Musto V, Mancini FP. Sirtuins as Mediator of the Anti-Ageing Effects of Calorie Restriction in Skeletal and Cardiac Muscle. Int J Mol Sci 2018; 19:E928. [PMID: 29561771 PMCID: PMC5979282 DOI: 10.3390/ijms19040928] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/14/2018] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
Fighting diseases and controlling the signs of ageing are the major goals of biomedicine. Sirtuins, enzymes with mainly deacetylating activity, could be pivotal targets of novel preventive and therapeutic strategies to reach such aims. Scientific proofs are accumulating in experimental models, but, to a minor extent, also in humans, that the ancient practice of calorie restriction could prove an effective way to prevent several degenerative diseases and to postpone the detrimental signs of ageing. In the present review, we summarize the evidence about the central role of sirtuins in mediating the beneficial effects of calorie restriction in skeletal and cardiac muscle since these tissues are greatly damaged by diseases and advancing years. Moreover, we entertain the possibility that the identification of sirtuin activators that mimic calorie restriction could provide the benefits without the inconvenience of this dietary style.
Collapse
Affiliation(s)
- Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
- CEINGE Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Emanuela Simone
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | - Maddalena Grimaldi
- Department of Pediatric Oncology and Hematology, Charité University Hospital, 13353 Berlin, Germany.
| | - Vincenzina Musto
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | | |
Collapse
|
38
|
D'Onofrio N, Servillo L, Balestrieri ML. SIRT1 and SIRT6 Signaling Pathways in Cardiovascular Disease Protection. Antioxid Redox Signal 2018; 28:711-732. [PMID: 28661724 PMCID: PMC5824538 DOI: 10.1089/ars.2017.7178] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Oxidative stress represents the common hallmark of pathological conditions associated with cardiovascular disease (CVD), including atherosclerosis, heart failure, hypertension, aging, diabetes, and other vascular system-related diseases. The sirtuin (SIRT) family, comprising seven proteins (SIRT1-SIRT7) sharing a highly conserved nicotinamide adenine dinucleotide (NAD+)-binding catalytic domain, attracted a great attention for the past few years as stress adaptor and epigenetic enzymes involved in the cellular events controlling aging-related disorder, cancer, and CVD. Recent Advances: Among sirtuins, SIRT1 and SIRT6 are the best characterized for their protective roles against inflammation, vascular aging, heart disease, and atherosclerotic plaque development. This latest role has been only recently unveiled for SIRT6. Of interest, in recent years, complex signaling networks controlled by SIRT1 and SIRT6 common to stress resistance, vascular aging, and CVD have emerged. CRITICAL ISSUES We provide a comprehensive overview of recent developments on the molecular signaling pathways controlled by SIRT1 and SIRT6, two post-translational modifiers proven to be valuable tools to dampen inflammation and oxidative stress at the cardiovascular level. FUTURE DIRECTIONS A deeper understanding of the epigenetic mechanisms through which SIRT1 and SIRT6 act in the signalings responsible for onset and development CVD is a prime scientific endeavor of the upcoming years. Multiple "omic" technologies will have widespread implications in understanding such mechanisms, speeding up the achievement of selective and efficient pharmacological modulation of sirtuins for future applications in the prevention and treatment of CVD. Antioxid. Redox Signal. 28, 711-732.
Collapse
Affiliation(s)
- Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, School of Medicine and Surgery, Università degli Studi della Campania , Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, School of Medicine and Surgery, Università degli Studi della Campania , Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, School of Medicine and Surgery, Università degli Studi della Campania , Naples, Italy
| |
Collapse
|
39
|
Pillai VB, Kanwal A, Fang YH, Sharp WW, Samant S, Arbiser J, Gupta MP. Honokiol, an activator of Sirtuin-3 (SIRT3) preserves mitochondria and protects the heart from doxorubicin-induced cardiomyopathy in mice. Oncotarget 2018; 8:34082-34098. [PMID: 28423723 PMCID: PMC5470953 DOI: 10.18632/oncotarget.16133] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/27/2017] [Indexed: 12/30/2022] Open
Abstract
Doxorubicin is the chemotherapeutic drug of choice for a wide variety of cancers, and cardiotoxicity is one of the major side effects of doxorubicin treatment. One of the main cellular targets of doxorubicin in the heart is mitochondria. Mitochondrial sirtuin, SIRT3 has been shown to protect against doxorubicin-induced cardiotoxicity. We have recently identified honokiol (HKL) as an activator of SIRT3, which protects the heart from developing pressure overload hypertrophy. Here, we show that HKL-mediated activation of SIRT3 also protects the heart from doxorubicin-induced cardiac damage without compromising the tumor killing potential of doxorubicin. Doxorubicin-induced cardiotoxicity is associated with increased ROS production and consequent fragmentation of mitochondria and cell death. HKL-mediated activation of SIRT3 prevented Doxorubicin induced ROS production, mitochondrial damage and cell death in rat neonatal cardiomyocytes. HKL also promoted mitochondrial fusion. We also show that treatment with HKL blocked doxorubicin-induced cardiac toxicity in mice. This was associated with reduced mitochondrial DNA damage and improved mitochondrial function. Furthermore, treatments of mice, bearing prostrate tumor-xenografts, with HKL and doxorubicin showed inhibition of tumor growth with significantly reduced cardiac toxicity. Our results suggest that HKL-mediated activation of SIRT3 protects the heart from doxorubicin-induced cardiotoxicity and represents a potentially novel adjunct for chemotherapy treatments.
Collapse
Affiliation(s)
- Vinodkumar B Pillai
- Department of Surgery, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Abhinav Kanwal
- Department of Surgery, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Yong Hu Fang
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Willard W Sharp
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Sadhana Samant
- Department of Surgery, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Jack Arbiser
- Department of Dermatology, Atlanta Veterans Administration Health Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Mahesh P Gupta
- Department of Surgery, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
40
|
Diguet N, Trammell SAJ, Tannous C, Deloux R, Piquereau J, Mougenot N, Gouge A, Gressette M, Manoury B, Blanc J, Breton M, Decaux JF, Lavery GG, Baczkó I, Zoll J, Garnier A, Li Z, Brenner C, Mericskay M. Nicotinamide Riboside Preserves Cardiac Function in a Mouse Model of Dilated Cardiomyopathy. Circulation 2017; 137:2256-2273. [PMID: 29217642 DOI: 10.1161/circulationaha.116.026099] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.
Collapse
Affiliation(s)
- Nicolas Diguet
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Samuel A J Trammell
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City (S.A.J.T., C.B.)
| | - Cynthia Tannous
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Robin Deloux
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | | | - Nathalie Mougenot
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Plateforme PECMV, UMS28, Paris, France (N.M.)
| | - Anne Gouge
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Mélanie Gressette
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Boris Manoury
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Jocelyne Blanc
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Marie Breton
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Jean-François Decaux
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, United Kingdom (G.G.L.)
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Hungary (I.B.)
| | - Joffrey Zoll
- Physiology Department, Faculty of Medicine and EA3072, Université de Strasbourg, France (J.Z.)
| | - Anne Garnier
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Zhenlin Li
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City (S.A.J.T., C.B.)
| | - Mathias Mericskay
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.).
| |
Collapse
|
41
|
Yu Y, Wang L, Delguste F, Durand A, Guilbaud A, Rousselin C, Schmidt AM, Tessier F, Boulanger E, Neviere R. Advanced glycation end products receptor RAGE controls myocardial dysfunction and oxidative stress in high-fat fed mice by sustaining mitochondrial dynamics and autophagy-lysosome pathway. Free Radic Biol Med 2017; 112:397-410. [PMID: 28826719 DOI: 10.1016/j.freeradbiomed.2017.08.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/05/2017] [Accepted: 08/17/2017] [Indexed: 12/18/2022]
Abstract
Oxidative stress and mitochondrial dysfunction are recognized as major contributors of cardiovascular damage in diabetes and high fat diet (HFD) fed mice. Blockade of receptor for advanced glycation end products (RAGE) attenuates vascular oxidative stress and development of atherosclerosis. We tested whether HFD-induced myocardial dysfunction would be reversed in RAGE deficiency mice, in association with changes in oxidative stress damage, mitochondrial respiration, mitochondrial fission and autophagy-lysosomal pathway. Cardiac antioxidant capacity was upregulated in RAGE-/- mice under normal diet as evidenced by increased superoxide dismutase and sirtuin mRNA expressions. Mitochondrial fragmentation and mitochondrial fission protein Drp1 and Fis1 expressions were increased in RAGE-/- mice. Autophagy-related protein expressions and cathepsin-L activity were increased in RAGE-/- mice suggesting sustained autophagy-lysosomal flux. HFD induced mitochondrial respiration defects, cardiac contractile dysfunction, disrupted mitochondrial dynamics and autophagy inhibition, which were partially prevented in RAGE-/- mice. Our results suggest that cardioprotection against HFD in RAGE-/- mice include reactivation of autophagy, as inhibition of autophagic flux by chloroquine fully abrogated beneficial myocardial effects and its stimulation by rapamycin improved myocardial function in HFD wild type mice. As mitochondrial fission is necessary to mitophagy, increased fragmentation of mitochondrial network in HFD RAGE-/- mice may have facilitated removal of damaged mitochondria leading to better mitochondrial quality control. In conclusion, modulation of RAGE pathway may improve mitochondrial damage and myocardial dysfunction in HFD mice. Attenuation of cardiac oxidative stress and maintenance of healthy mitochondria population ensuring adequate energy supply may be involved in myocardial protection against HFD.
Collapse
Affiliation(s)
- Yichi Yu
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France; School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Wang
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France; School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Florian Delguste
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France
| | - Arthur Durand
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France; University Hospital CHU of Lille, F-59000 Lille, France
| | - Axel Guilbaud
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France
| | - Clementine Rousselin
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France; University Hospital CHU of Lille, F-59000 Lille, France
| | - Ann Marie Schmidt
- Diabetes Research Center, Department of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Frédéric Tessier
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France
| | - Eric Boulanger
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France; University Hospital CHU of Lille, F-59000 Lille, France
| | - Remi Neviere
- INSERM U995, LIRIC /Team "Glycation: from inflammation to aging", Lille University, France; University Hospital CHU of Martinique, University of Antilles, Fort de France F-97200, France.
| |
Collapse
|
42
|
Sosnowska B, Mazidi M, Penson P, Gluba-Brzózka A, Rysz J, Banach M. The sirtuin family members SIRT1, SIRT3 and SIRT6: Their role in vascular biology and atherogenesis. Atherosclerosis 2017; 265:275-282. [DOI: 10.1016/j.atherosclerosis.2017.08.027] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022]
|
43
|
Feng GS, Zhu CG, Li ZM, Wang PX, Huang Y, Liu M, He P, Lou LL, Chen SR, Liu PQ. Synthesis of the novel PARP-1 inhibitor AG-690/11026014 and its protective effects on angiotensin II-induced mouse cardiac remodeling. Acta Pharmacol Sin 2017; 38:638-650. [PMID: 28239158 DOI: 10.1038/aps.2016.159] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/24/2016] [Indexed: 12/12/2022] Open
Abstract
We previously identified AG-690/11026014 (6014) as a novel poly(ADP-ribose) polymerase-1 (PARP-1) inhibitor that effectively prevented angiotensin II (Ang II)-induced cardiomyocyte hypertrophy. In the present study, we reported a new synthesis route for 6014, and investigated its protective effects on Ang II-induced cardiac remodeling and cardiac dysfunction and the underlying mechanisms in mice. We designed a new synthesis route to obtain a sufficient quantity of 6014 for this in vivo study. C57BL/6J mice were infused with Ang II and treated with 6014 (10, 30, 90 mg·kg-1·d-1, ig) for 4 weeks. Then two-dimensional echocardiography was performed to assess the cardiac function and structure. Histological changes of the hearts were examined with HE staining and Masson's trichrome staining. The protein expression was evaluated by Western blot, immunohistochemistry and immunofluorescence assays. The activities of sirtuin-1 (SIRT-1) and the content of NAD+ were detected with the corresponding test kits. Treatment with 6014 dose-dependently improved cardiac function, including LVEF, CO and SV and reversed the changes of cardiac structure in Ang II-infused mice: it significantly ameliorated Ang II-induced cardiac hypertrophy evidenced by attenuating the enlargement of cardiomyocytes, decreased HW/BW and LVW/BW, and decreased expression of hypertrophic markers ANF, BNP and β-MHC; it also prevented Ang II-induced cardiac fibrosis, as implied by the decrease in excess accumulation of extracellular matrix (ECM) components collagen I, collagen III and FN. Further studies revealed that treatment with 6014 did not affect the expression levels of PARP-1, but dose-dependently inhibited the activity of PARP-1 and subsequently restored the activity of SIRT-1 in heart tissues due to the decreased consumption of NAD+ and attenuated Poly-ADP-ribosylation (PARylation) of SIRT-1. In conclusion, the novel PARP-1 inhibitor 6014 effectively protects mice against AngII-induced cardiac remodeling and improves cardiac function. Thus, 6014 might be a potential therapeutic agent for heart diseases..
Collapse
|
44
|
Vitiello M, Zullo A, Servillo L, Mancini FP, Borriello A, Giovane A, Della Ragione F, D'Onofrio N, Balestrieri ML. Multiple pathways of SIRT6 at the crossroads in the control of longevity, cancer, and cardiovascular diseases. Ageing Res Rev 2017; 35:301-311. [PMID: 27829173 DOI: 10.1016/j.arr.2016.10.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 12/27/2022]
Abstract
Sirtuin 6 (SIRT6) is a member of the sirtuin family NAD+-dependent deacetylases with multiple roles in controlling organism homeostasis, lifespan, and diseases. Due to its complex and opposite functional roles, this sirtuin is considered a two-edged sword in health and disease. Indeed, SIRT6 improves longevity, similarly to the founding yeast member, silent information regulator-2 (Sir2), and modulates genome stability, telomere integrity, transcription, and DNA repair. Its deficiency is associated with chronic inflammation, diabetes, cardiac hypertrophy, obesity, liver dysfunction, muscle/adipocyte disorders, and cancer. Besides, pieces of evidence showed that SIRT6 is a promoter of specific oncogenic pathways, thus disclosing its dual role regarding cancer development. Collectively, these findings suggest that multiple mechanisms, to date not entirely known, underlie the intriguing roles of SIRT6. Here we provide an overview of the current molecular mechanisms through which SIRT6 controls cancer and heart diseases, and describe its recent implications in the atherosclerotic plaque development.
Collapse
Affiliation(s)
- Milena Vitiello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy; CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | | | - Adriana Borriello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alfonso Giovane
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.
| |
Collapse
|
45
|
Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders - A step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1066-1077. [PMID: 27836629 PMCID: PMC5423868 DOI: 10.1016/j.bbadis.2016.11.010] [Citation(s) in RCA: 832] [Impact Index Per Article: 118.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 01/06/2023]
Abstract
Mitochondria are the powerhouses of the cell and are involved in essential functions of the cell, including ATP production, intracellular Ca2+ regulation, reactive oxygen species production & scavenging, regulation of apoptotic cell death and activation of the caspase family of proteases. Mitochondrial dysfunction and oxidative stress are largely involved in aging, cancer, age-related neurodegenerative and metabolic syndrome. In the last decade, tremendous progress has been made in understanding mitochondrial structure, function and their physiology in metabolic syndromes such as diabetes, obesity, stroke and hypertension, and heart disease. Further, progress has also been made in developing therapeutic strategies, including lifestyle interventions (healthy diet and regular exercise), pharmacological strategies and mitochondria-targeted approaches. These strategies were mainly focused to reduce mitochondrial dysfunction and oxidative stress and to maintain mitochondrial quality in metabolic syndromes. The purpose of our article is to highlight the recent progress on the mitochondrial role in metabolic syndromes and also summarize the progress of mitochondria-targeted molecules as therapeutic targets to treat metabolic syndromes. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Department of Biotechnology and Bioinformatics, Sri Guru Gobind Singh College, Sector-26, Chandigarh 160019, India; Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Gurjit Kaur Bhatti
- UGC Centre of Excellence in Nano applications, Panjab University, UIPS building, Chandigarh 160014, India
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neuroscience & Pharmacology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neurology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, MS 7495, Lubbock, TX 79413, United States
| |
Collapse
|
46
|
Loria-Kohen V, Marcos-Pasero H, de la Iglesia R, Aguilar-Aguilar E, Espinosa-Salinas I, Herranz J, Ramírez de Molina A, Reglero G. Multiple chemical sensitivity: Genotypic characterization, nutritional status and quality of life in 52 patients. Med Clin (Barc) 2017; 149:141-146. [PMID: 28283271 DOI: 10.1016/j.medcli.2017.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/21/2016] [Accepted: 01/17/2017] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVES Multiple chemical sensitivity (MCS) is a chronic, multisystem syndrome of unknown etiology. The aim of the present study was to describe the nutritional status and quality of life of patients suffering from MCS, as well as to identify potential polymorphisms associated with this illness. PATIENTS AND METHODS A cross-sectional, descriptive study was performed on patients with a diagnosis of MCS. Data on anthropometric and body composition variables, hand muscle strength and quality of life were collected. The selection of single nucleotide polymorphisms (SNPs) was based on genes previously associated with MCS and genes involved in inflammatory and oxidative stress pathways. RESULTS A total of 52 patients (93.2% female), with a mean age of 50.9 (10.3) years were included in the study. Among them, based on their BMI, 48% had an inadequate nutritional status (17% were underweight and 32% were overweight or obese). Thirty percent of patients had a low muscle mass for their age, 84% had muscle strength below the tenth percentile, and 51.8% had a high fat mass percentage. Regarding quality of life, all median scores were lower than those of other illnesses assessed for every subscale assessed. Statistically significant differences between patient cases and controls were found with respect to rs1801133 (MTHFR), rs174546 (FADS1) and rs1801282 (PPARγ) polymorphisms. CONCLUSION A high percentage of patients had a poor nutritional status, low muscle strength and decreased muscle mass. These facts exacerbate the already-lower quality of life of these patients. Specific genetic polymorphisms associated with the syndrome or its pathogenesis were not identified.
Collapse
Affiliation(s)
- Viviana Loria-Kohen
- IMDEA-Food, Campus de Excelencia Internacional (CEI) UAM+CSIC, Madrid, España.
| | | | - Rocío de la Iglesia
- IMDEA-Food, Campus de Excelencia Internacional (CEI) UAM+CSIC, Madrid, España
| | | | | | - Jesús Herranz
- IMDEA-Food, Campus de Excelencia Internacional (CEI) UAM+CSIC, Madrid, España
| | | | - Guillermo Reglero
- IMDEA-Food, Campus de Excelencia Internacional (CEI) UAM+CSIC, Madrid, España; Departamento de Producción y Caracterización de Nuevos Alimentos, Instituto de Investigación en Ciencias de la Alimentación (CIAL), Campus de Excelencia Internacional (CEI) UAM+CSIC, Madrid, España
| |
Collapse
|
47
|
Bhatti JS, Kumar S, Vijayan M, Bhatti GK, Reddy PH. Therapeutic Strategies for Mitochondrial Dysfunction and Oxidative Stress in Age-Related Metabolic Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 146:13-46. [PMID: 28253984 DOI: 10.1016/bs.pmbts.2016.12.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondria are complex, intercellular organelles present in the cells and are involved in multiple roles including ATP formation, free radicals generation and scavenging, calcium homeostasis, cellular differentiation, and cell death. Many studies depicted the involvement of mitochondrial dysfunction and oxidative damage in aging and pathogenesis of age-related metabolic disorders and neurodegenerative diseases. Remarkable advancements have been made in understanding the structure, function, and physiology of mitochondria in metabolic disorders such as diabetes, obesity, cardiovascular diseases, and stroke. Further, much progress has been done in the improvement of therapeutic strategies, including lifestyle interventions, pharmacological, and mitochondria-targeted therapeutic approaches. These strategies were mainly focused to reduce the mitochondrial dysfunction caused by oxidative stress and to retain the mitochondrial health in various diseases. In this chapter, we have highlighted the involvement of mitochondrial dysfunction in the pathophysiology of various disorders and recent progress in the development of mitochondria-targeted molecules as therapeutic measures for metabolic disorders.
Collapse
Affiliation(s)
- J S Bhatti
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India.
| | - S Kumar
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - M Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - G K Bhatti
- UGC Centre of Excellence in Nano Applications, Panjab University, Chandigarh, India
| | - P H Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
48
|
Lin Q, Geng Y, Lin S, Tian Z. Sirtuin1 (SIRT1) Regulates Tumor Necrosis Factor-alpha (TNF-α-Induced) Aquaporin-2 (AQP2) Expression in Renal Medullary Collecting Duct Cells Through Inhibiting the NF-κB Pathway. Med Sci Monit Basic Res 2016; 22:165-174. [PMID: 27980322 PMCID: PMC5189724 DOI: 10.12659/msmbr.901909] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Aquaporin-2 (AQP2) plays a major role in water reabsorption in the renal collecting duct, and is involved in a variety of renal disease. Recent studies have indicate that sirtuin1 (SIRT1) exerts renoprotective properties against kidney diseases. This study aimed to determine the potential role of SIRT1 in AQP2 expression induced by tumor necrosis factor-alpha (TNF-α) and to disclose the underlying mechanism in renal inner medullary collecting duct (IMCD) cells. Material/Methods Quantitative real-time PCR and Western blotting were respectively identified mRNA and protein expression. Immunofluorescence staining was used to detect the localization of AQP2. Small-interfering RNA (siRNA) was carried out for mechanism study. Results Results showed that AQP2 was clearly increased in the plasma membrane and decreased in the cytoplasm of IMCD cells treated with AVP. TNF-α treatment in IMCD cells significantly reduced SIRT1 and AQP2 expression, and increased acetylated NF-κBp65 protein level in time- and concentration-dependent manners. Moreover, SIRT1 overexpression or the activator SRT1720 augmented AQP2 expression and reduced the acetylation of NF-κBp65, which was reversed by SIRT1 siRNA or the inhibitors Ex527 and sirtinol in TNF-α-induced IMCD cells. Knockdown of NF-κBp65 or NF-κBp65 inhibition by pyrrolidine dithiocarbamate (PDTC) enhanced AQP2 expression in IMCD cells exposed to TNF-α. Importantly, knockdown of NF-κBp65 augmented the up-regulation of SIRT1 on AQP2 expression in IMCD cells induced by TNF-α. Conclusions These findings indicate that SIRT1 increases AQP2 expression in TNF-α-induced IMCD cells via the NF-κB-dependent signalling pathway, which might provide novel insight to understanding the renoprotective effects of SIRT1 in kidney diseases.
Collapse
Affiliation(s)
- Qinqin Lin
- College of Physical Education, Yanshan University, Qinhuangdao, Hebei, China (mainland)
| | - Yuanwen Geng
- College of Physical Education, Yanshan University, Qinhuangdao, Hebei, China (mainland)
| | - Shuaishuai Lin
- Department of Education, Beijing Sport University, Beijing, China (mainland)
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
49
|
Ajami M, Pazoki-Toroudi H, Amani H, Nabavi SF, Braidy N, Vacca RA, Atanasov AG, Mocan A, Nabavi SM. Therapeutic role of sirtuins in neurodegenerative disease and their modulation by polyphenols. Neurosci Biobehav Rev 2016; 73:39-47. [PMID: 27914941 DOI: 10.1016/j.neubiorev.2016.11.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/26/2016] [Accepted: 11/26/2016] [Indexed: 12/15/2022]
Abstract
Searching for effective therapeutic agents to prevent neurodegeneration is a challenging task due to the growing list of neurodegenerative disorders associated with a multitude of inter-related pathways. The induction and inhibition of several different signaling pathways has been shown to slow down and/or attenuate neurodegeneration and decline in cognition and locomotor function. Among these signaling pathways, a new class of enzymes known as sirtuins or silent information regulators of gene transcription has been shown to play important regulatory roles in the ageing process. SIRT1, a nuclear sirtuin, has received particular interest due to its role as a deacetylase for several metabolic and signaling proteins involved in stress response, apoptosis, mitochondrial function, self-renewal, and neuroprotection. A new strategy to treat neurodegenerative diseases is targeted therapy. In this paper, we reviewed up-to-date findings regarding the targeting of SIRT1 by polyphenolic compounds, as a new approach in the search for novel, safe and effective treatments for neurodegenerative diseases. .
Collapse
Affiliation(s)
- Marjan Ajami
- National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Pazoki-Toroudi
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Amani
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Australia.
| | - Rosa Anna Vacca
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari, Italy.
| | - Atanas Georgiev Atanasov
- Department of Pharmacognosy, University of Vienna, 1090 Vienna, Austria; Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Andrei Mocan
- Department of Pharmaceutical Botany, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Xu S, Bai P, Jin ZG. Sirtuins in Cardiovascular Health and Diseases. Trends Endocrinol Metab 2016; 27:677-678. [PMID: 27461006 PMCID: PMC5995322 DOI: 10.1016/j.tem.2016.07.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14640, USA
| | - Peter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Lendület Laboratory of Cellular Metabolism Research Group, Debrecen, Hungary; Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14640, USA.
| |
Collapse
|