101
|
Wagner DR, Karnik S, Gunderson ZJ, Nielsen JJ, Fennimore A, Promer HJ, Lowery JW, Loghmani MT, Low PS, McKinley TO, Kacena MA, Clauss M, Li J. Dysfunctional stem and progenitor cells impair fracture healing with age. World J Stem Cells 2019; 11:281-296. [PMID: 31293713 PMCID: PMC6600851 DOI: 10.4252/wjsc.v11.i6.281] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/26/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Successful fracture healing requires the simultaneous regeneration of both the bone and vasculature; mesenchymal stem cells (MSCs) are directed to replace the bone tissue, while endothelial progenitor cells (EPCs) form the new vasculature that supplies blood to the fracture site. In the elderly, the healing process is slowed, partly due to decreased regenerative function of these stem and progenitor cells. MSCs from older individuals are impaired with regard to cell number, proliferative capacity, ability to migrate, and osteochondrogenic differentiation potential. The proliferation, migration and function of EPCs are also compromised with advanced age. Although the reasons for cellular dysfunction with age are complex and multidimensional, reduced expression of growth factors, accumulation of oxidative damage from reactive oxygen species, and altered signaling of the Sirtuin-1 pathway are contributing factors to aging at the cellular level of both MSCs and EPCs. Because of these geriatric-specific issues, effective treatment for fracture repair may require new therapeutic techniques to restore cellular function. Some suggested directions for potential treatments include cellular therapies, pharmacological agents, treatments targeting age-related molecular mechanisms, and physical therapeutics. Advanced age is the primary risk factor for a fracture, due to the low bone mass and inferior bone quality associated with aging; a better understanding of the dysfunctional behavior of the aging cell will provide a foundation for new treatments to decrease healing time and reduce the development of complications during the extended recovery from fracture healing in the elderly.
Collapse
Affiliation(s)
- Diane R Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States
| | - Alanna Fennimore
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Hunter J Promer
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - M Terry Loghmani
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| | - Matthias Clauss
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jiliang Li
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| |
Collapse
|
102
|
Mellini P, Itoh Y, Elboray EE, Tsumoto H, Li Y, Suzuki M, Takahashi Y, Tojo T, Kurohara T, Miyake Y, Miura Y, Kitao Y, Kotoku M, Iida T, Suzuki T. Identification of Diketopiperazine-Containing 2-Anilinobenzamides as Potent Sirtuin 2 (SIRT2)-Selective Inhibitors Targeting the "Selectivity Pocket", Substrate-Binding Site, and NAD +-Binding Site. J Med Chem 2019; 62:5844-5862. [PMID: 31144814 DOI: 10.1021/acs.jmedchem.9b00255] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The NAD+-dependent deacetylase SIRT2 represents an attractive target for drug development. Here, we designed and synthesized drug-like SIRT2-selective inhibitors based on an analysis of the putative binding modes of recently reported SIRT2-selective inhibitors and evaluated their SIRT2-inhibitory activity. This led us to develop a more drug-like diketopiperazine structure as a "hydrogen bond (H-bond) hunter" to target the substrate-binding site of SIRT2. Thioamide 53, a conjugate of diketopiperazine and 2-anilinobenzamide which is expected to occupy the "selectivity pocket" of SIRT2, exhibited potent SIRT2-selective inhibition. Inhibition of SIRT2 by 53 was mediated by the formation of a 53-ADP-ribose conjugate, suggesting that 53 is a mechanism-based inhibitor targeting the "selectivity pocket", substrate-binding site, and NAD+-binding site. Furthermore, 53 showed potent antiproliferative activity toward breast cancer cells and promoted neurite outgrowth of Neuro-2a cells. These findings should pave the way for the discovery of novel therapeutic agents for cancer and neurological disorders.
Collapse
Affiliation(s)
- Paolo Mellini
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Yukihiro Itoh
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Elghareeb E Elboray
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan.,Chemistry Department, Faculty of Science , South Valley University , Qena 83523 , Egypt
| | - Hiroki Tsumoto
- Research Team for Mechanism of Aging , Tokyo Metropolitan Institute of Gerontology , 35-2 Sakae-cho , Itabashi-ku, Tokyo 173-0015 , Japan
| | - Ying Li
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Miki Suzuki
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Yukari Takahashi
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Toshifumi Tojo
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Takashi Kurohara
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Yuka Miyake
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging , Tokyo Metropolitan Institute of Gerontology , 35-2 Sakae-cho , Itabashi-ku, Tokyo 173-0015 , Japan
| | - Yuki Kitao
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Masayuki Kotoku
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Tetsuya Iida
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan
| | - Takayoshi Suzuki
- Graduate School of Medical Science , Kyoto Prefectural University of Medicine , 1-5 Shimogamohangi-cho , Sakyo-ku, Kyoto 606-0823 , Japan.,CREST , Japan Science and Technology Agency (JST) , 4-1-8 Honcho Kawaguchi , Saitama 332-0012 , Japan
| |
Collapse
|
103
|
Ren X, Chen N, Chen Y, Liu W, Hu Y. TRB3 stimulates SIRT1 degradation and induces insulin resistance by lipotoxicity via COP1. Exp Cell Res 2019; 382:111428. [PMID: 31125554 DOI: 10.1016/j.yexcr.2019.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/05/2019] [Accepted: 05/08/2019] [Indexed: 11/26/2022]
Abstract
Fatty acid-induced lipotoxicity plays an important role in the pathogenesis of diabetes mellitus. Our previous studies have documented that lipotoxicity contributes to the onset and development of diabetes via insulin resistance and/or compromised function of the pancreatic β-cells. However, the underlying molecular mechanisms associating lipotoxicity with insulin resistance remain to be fully elucidated. In this study, we explored the role of TRB3-COP1-SIRT1 in lipotoxicity leading to insulin resistance in hepatocytes. High fat diet (HFD)-fed mice and hepG2 cells stimulated with palmitate were utilized as models of lipid metabolism disorders. We analyzed the interactions of SIRT1 and COP1 with each other and with TRB3 using co-immunoprecipitation, western blotting. SIRT1 ubiquitination was also explored. Animal and cell experiments showed that lipotoxicity induced SIRT1 down-regulation at the protein level without altering the mRNA level, whereas, lipotoxicity led to up-regulation of TRB3 and COP1 at both the gene and protein levels. Mechanistic analysis indicated that COP1 functioned as an E3 Ub-ligase of SIRT1, responsible for its proteasomal degradation under lipotoxic conditions. TRB3 recruited COP1 to SIRT1 to promote its ubiquitination. Our data indicated for the first time that TRB3-COP1-SIRT1 pathway played an important role in lipotoxicity leading to insulin resistance in hepatocytes, and suggested that COP1 could be a potential therapeutic choice for the treatment of diabetes mellitus, with lipotoxicity being the important pathomechanism.
Collapse
Affiliation(s)
- Xingxing Ren
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Ningxin Chen
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Yawen Chen
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Wei Liu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| | - Yaomin Hu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| |
Collapse
|
104
|
Malde AK, Hill TA, Iyer A, Fairlie DP. Crystal Structures of Protein-Bound Cyclic Peptides. Chem Rev 2019; 119:9861-9914. [DOI: 10.1021/acs.chemrev.8b00807] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alpeshkumar K. Malde
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Timothy A. Hill
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Abishek Iyer
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David P. Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
105
|
Yu S, Zhou R, Yang T, Liu S, Cui Z, Qiao Q, Zhang J. Hypoxia promotes colorectal cancer cell migration and invasion in a SIRT1-dependent manner. Cancer Cell Int 2019; 19:116. [PMID: 31068761 PMCID: PMC6492435 DOI: 10.1186/s12935-019-0819-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022] Open
Abstract
Background Hypoxic microenvironments play a significant role in the progression of colorectal cancer (CRC). Silencing information regulator 1 (SIRT1), a class III histone deacetylase, modulates the multiple biological behaviors of cancer. However, its role in CRC remains unclear. This study aims to explore the role of SIRT1 in CRC migration and invasion under hypoxia. Methods SIRT1 protein and mRNA levels were detected by Western blotting and real-time PCR in CRC cells exposed to hypoxia (1% O2). The migration and invasion abilities of SW480 and HCT116 cells with SIRT1 overexpression or knockdown were studied with transwell assays, and the results were confirmed by those of treatment with specific SIRT1 activator (SRT1720) and inhibitor (EX527). The dual-luciferase reporter systems with a series of SIRT1 promoter truncations were used to analyze their transcriptional activities, respectively. After a bioinformatic analysis of potential transcription factors, the direct interaction between the transcription factor and SIRT1 promoter was determined by chromatin immunoprecipitation (ChIP) assays. Western blot and real-time PCR assays were used to detect the activation and acetylation levels of the NF-κB pathway. Results The protein and mRNA levels of SIRT1 were significantly decreased under hypoxia, and these effects were replicated by cobalt chloride treatment. Hypoxia promoted cell migration and invasion, which were impeded by the overexpression or activation of SIRT1 and promoted by the knockdown or inhibition of SIRT1. The dual-luciferase reporter gene and ChIP analyses revealed that the core regulatory elements located 100 bp upstream of the SIRT1 promoter and early growth response factor 1 (EGR1) could interact with this DNA sequence. Subsequent rescue experiments suggested that EGR1 was essential for hypoxia-mediated SIRT1 transcriptional suppression. Western blot analyses demonstrated that SIRT1 overexpression eliminated the p65 acetylation induced by hypoxia along with the decreased MMP-2/-9, suggesting that NF-κB was a direct downstream target of SIRT1 and might regulate cell migration and invasion through MMP-2/-9. Conclusions Our results establish for the first time that EGR1 plays an important role in regulating SIRT1 expression under hypoxia. Hypoxia promotes CRC cell migration and invasion in a SIRT1-dependent manner. And a potential SIRT1/NF-κB/MMP-2/-9 axis modulates this process. Electronic supplementary material The online version of this article (10.1186/s12935-019-0819-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shentong Yu
- 1State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032 China
| | - Ru Zhou
- 1State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032 China
| | - Tong Yang
- 1State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032 China
| | - Shuang Liu
- 2School of Basic Medicine, The Fourth Military Medical University, Xi'an, 710032 China
| | - Zhuqing Cui
- 1State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032 China
| | - Qing Qiao
- 3Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| | - Jing Zhang
- 1State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032 China
| |
Collapse
|
106
|
Abstract
The sirtuin family of NAD+-dependent protein deacetylases promotes longevity and counteracts age-related diseases. One of the major targets of Sirtuins are the FoxO family of transcription factors. FoxOs play a major role in the adaptation of cells to a variety of stressors such as oxidative stress and growth factor deprivation. Studies with murine models of cell-specific loss- or gain-of-function of Sirtuins or FoxOs and with Sirtuin1 stimulators have provided novel insights into the function and signaling of these proteins on the skeleton. These studies have revealed that both Sirtuins and FoxOs acting directly in cartilage and bone cells are critical for normal skeletal development, homeostasis and that their dysregulation might contribute to skeletal disease. Deacetylation of FoxOs by Sirt1 in osteoblasts and osteoclasts stimulates bone formation and inhibits bone resorption, making Sirt1 ligands promising therapeutic agents for diseases of low bone mass. While a similar link has not been established in chondrocytes, Sirt1 and FoxOs both have chondroprotective actions, suggesting that Sirt1 activators may have similar efficacy in preventing cartilage degeneration due to aging or injury. In this review we summarize these advances and discuss their implications for the pathogenesis of age-related osteoporosis and osteoarthritis.
Collapse
Affiliation(s)
- Maria Almeida
- Department of Medicine, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Orthopedics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Ryan M Porter
- Department of Medicine, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Orthopedics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
107
|
Dembic M, Andersen HS, Bastin J, Doktor TK, Corydon TJ, Sass JO, Lopes Costa A, Djouadi F, Andresen BS. Next generation sequencing of RNA reveals novel targets of resveratrol with possible implications for Canavan disease. Mol Genet Metab 2019; 126:64-76. [PMID: 30446350 DOI: 10.1016/j.ymgme.2018.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022]
Abstract
Resveratrol (RSV) is a small compound first identified as an activator of sirtuin 1 (SIRT1), a key factor in mediating the effects of caloric restriction. Since then, RSV received great attention for its widespread beneficial effects on health and in connection to many diseases. RSV improves the metabolism and the mitochondrial function, and more recently it was shown to restore fatty acid β-oxidation (FAO) capacities in patient fibroblasts harboring mutations with residual enzyme activity. Many of RSV's beneficial effects are mediated by the transcriptional coactivator PGC-1α, a direct target of SIRT1 and a master regulator of the mitochondrial fatty acid oxidation. Despite numerous studies RSV's mechanism of action is still not completely elucidated. Our aim was to investigate the effects of RSV on gene regulation on a wide scale, possibly to detect novel genes whose up-regulation by RSV may be of interest with respect to disease treatment. We performed Next Generation Sequencing of RNA on normal fibroblasts treated with RSV. To investigate whether the effects of RSV are mediated through SIRT1 we expanded the analysis to include SIRT1-knockdown fibroblasts. We identified the aspartoacylase (ASPA) gene, mutated in Canavan disease, to be strongly up-regulated by RSV in several cell lines, including Canavan disease fibroblasts. We further link RSV to the up-regulation of other genes involved in myelination including the glial specific transcription factors POU3F1, POU3F2, and myelin basic protein (MBP). We also observe a strong up-regulation by RSV of the riboflavin transporter gene SLC52a1. Mutations in SLC52a1 cause transient multiple acyl-CoA dehydrogenase deficiency (MADD). Our analysis of alternative splicing identified novel metabolically important genes affected by RSV, among which is particularly interesting the α subunit of the stimulatory G protein (Gsα), which regulates the cellular levels of cAMP through adenylyl cyclase. We conclude that in fibroblasts RSV stimulates the PGC-1α and p53 pathways, and up-regulates genes affecting the glucose metabolism, mitochondrial β-oxidation, and mitochondrial biogenesis. We further confirm that RSV might be a relevant treatment in the correction of FAO deficiencies and we suggest that treatment in other metabolic disorders including Canavan disease and MADD might be also beneficial.
Collapse
Affiliation(s)
- Maja Dembic
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Henriette S Andersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Jean Bastin
- INSERM UMR-S 1124, Université Paris Descartes, UFR Biomédicale des Saints-Pères, 45, rue des Saints-Pères, 75270 Paris, cedex 06, France
| | - Thomas K Doktor
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, 8000 Aarhus C, Denmark.
| | - Jörn Oliver Sass
- Research Group Inborn Errors of Metabolism, Department of Natural Sciences & IFGA, University of Applied Sciences, Rheinbach, Germany.
| | - Alexandra Lopes Costa
- INSERM UMR-S 1124, Université Paris Descartes, UFR Biomédicale des Saints-Pères, 45, rue des Saints-Pères, 75270 Paris, cedex 06, France
| | - Fatima Djouadi
- INSERM UMR-S 1124, Université Paris Descartes, UFR Biomédicale des Saints-Pères, 45, rue des Saints-Pères, 75270 Paris, cedex 06, France
| | - Brage S Andresen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
108
|
Ma L, Niu H, Sha G, Zhang Y, Liu P, Li Y. Serum SIRT1 Is Associated with Frailty and Adipokines in Older Adults. J Nutr Health Aging 2019; 23:246-250. [PMID: 30820512 DOI: 10.1007/s12603-018-1149-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Because frailty is a major health concern among older patients, identifying frailty-related biomarkers will help in the early detection and prevention of frailty. Thus, we aimed to determine the association between circulating levels of silent mating-type information regulation 2 homolog 1 (SIRT1) and frailty. METHODS We assessed circulating SIRT1 levels in 16 robust, 74 prefrail, and 40 frail older adults. Frailty was diagnosed based on the Fried phenotype. Circulating cytokine and adipokine (e.g., vaspin, adiponectin, and leptin) levels were assessed. Differences in SIRT1 levels among the three subject groups were compared; correlations of SIRT1 levels with physical function and adipokine and cytokine levels were analyzed. RESULTS Serum SIRT1 levels were significantly higher among frail older adults than among robust ones. Older adults with slowness or weight loss had high SIRT1 levels. Serum SIRT1 levels negatively correlated with gait speed, even after adjustment for age and sex; age; and insulin, vaspin, and leptin levels; they correlated negatively with phospholipase A2 levels. CONCLUSIONS High SIRT1 levels were observed in frail elderly patients and were correlated with decreased physical function. Insulin and adipokine levels might be the link between SIRT1 and frailty, whereas inflammation may not be involved in this process.
Collapse
Affiliation(s)
- L Ma
- Dr. Lina Ma, Department of Geriatrics, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing 100053, China. E-mail:
| | | | | | | | | | | |
Collapse
|
109
|
Krzysiak TC, Thomas L, Choi YJ, Auclair S, Qian Y, Luan S, Krasnow SM, Thomas LL, Koharudin LMI, Benos PV, Marks DL, Gronenborn AM, Thomas G. An Insulin-Responsive Sensor in the SIRT1 Disordered Region Binds DBC1 and PACS-2 to Control Enzyme Activity. Mol Cell 2018; 72:985-998.e7. [PMID: 30415949 PMCID: PMC6309500 DOI: 10.1016/j.molcel.2018.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/13/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022]
Abstract
Current models of SIRT1 enzymatic regulation primarily consider the effects of fluctuating levels of its co-substrate NAD+, which binds to the stably folded catalytic domain. By contrast, the roles of the sizeable disordered N- and C-terminal regions of SIRT1 are largely unexplored. Here we identify an insulin-responsive sensor in the SIRT1 N-terminal region (NTR), comprising an acidic cluster (AC) and a 3-helix bundle (3HB), controlling deacetylase activity. The allosteric assistor DBC1 removes a distal N-terminal shield from the 3-helix bundle, permitting PACS-2 to engage the acidic cluster and the transiently exposed helix 3 of the 3-helix bundle, disrupting its structure and inhibiting catalysis. The SIRT1 activator (STAC) SRT1720 binds and stabilizes the 3-helix bundle, protecting SIRT1 from inhibition by PACS-2. Identification of the SIRT1 insulin-responsive sensor and its engagement by the DBC1 and PACS-2 regulatory hub provides important insight into the roles of disordered regions in enzyme regulation and the mode by which STACs promote metabolic fitness.
Collapse
Affiliation(s)
- Troy C Krzysiak
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Laurel Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - You-Jin Choi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Sylvain Auclair
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Yiqi Qian
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Shan Luan
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Stephanie M Krasnow
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Laura L Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Leonardus M I Koharudin
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Daniel L Marks
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Angela M Gronenborn
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA; Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
110
|
Huang B, Zhang W, Wei L, Chen S, Wang T, Fu R. Resveratrol down-regulates endothelin type B receptors in vascular smooth muscle cells via Sirt1/ERK1/2/NF-кB signaling pathways. Eur J Pharmacol 2018; 840:44-49. [DOI: 10.1016/j.ejphar.2018.09.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 09/11/2018] [Accepted: 09/18/2018] [Indexed: 10/28/2022]
|
111
|
Suades R, Cosentino F. Sirtuin 1/soluble guanylyl cyclase: a nitric oxide-independent pathway to rescue ageing-induced vascular dysfunction. Cardiovasc Res 2018; 115:485-487. [DOI: 10.1093/cvr/cvy297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Rosa Suades
- Cardiology Unit, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Solna, 171 76 Stockholm, Sweden
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Solna, 171 76 Stockholm, Sweden
| |
Collapse
|
112
|
Xia H, Liu C, Li CC, Fu M, Takahashi S, Hu KQ, Aizawa K, Hiroyuki S, Wu G, Zhao L, Wang XD. Dietary Tomato Powder Inhibits High-Fat Diet-Promoted Hepatocellular Carcinoma with Alteration of Gut Microbiota in Mice Lacking Carotenoid Cleavage Enzymes. Cancer Prev Res (Phila) 2018; 11:797-810. [PMID: 30446518 DOI: 10.1158/1940-6207.capr-18-0188] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/28/2018] [Accepted: 10/02/2018] [Indexed: 11/16/2022]
Abstract
Both incidence and death rate due to liver cancer have increased in the United States. Higher consumption of lycopene-rich tomato and tomato products is associated with a decreased risk of cancers. β-Carotene-15, 15'-oxygenase (BCO1), and β-carotene-9', 10'-oxygenase (BCO2) cleave lycopene to produce bioactive apo-lycopenoids. Although BCO1/BCO2 polymorphisms affect human and animal lycopene levels, whether dietary tomato consumption can inhibit high-fat diet (HFD)-promoted hepatocellular carcinoma (HCC) development and affect gut microbiota in the absence of BCO1/BCO2 is unclear. BCO1/BCO2 double knockout mice were initiated with a hepatic carcinogen (diethylnitrosamine) at 2 weeks of age. At 6 weeks of age, the mice were randomly assigned to an HFD (60% of energy as fat) with or without tomato powder (TP) feeding for 24 weeks. Results showed that TP feeding significantly decreased HCC development (67%, 83%, and 95% reduction in incidence, multiplicity, and tumor volume, respectively, P < 0.05). Protective effects of TP feeding were associated with (1) decreased hepatic inflammatory foci development and mRNA expression of proinflammatory biomarkers (IL1β, IL6, IL12α, monocyte chemoattractant protein-1, and inducible NO synthase); (2) increased mRNA expression of deacetylase sirtuin 1 and nicotinamide phosphoribosyltransferase involving NAD+ production; and (3) increased hepatic circadian clock genes (circadian locomotor output cycles kaput, period 2, and cryptochrome-2, Wee1). Furthermore, TP feeding increased gut microbial richness and diversity, and significantly decreased the relative abundance of the genus Clostridium and Mucispirillum, respectively. The present study demonstrates that dietary tomato feeding independent of carotenoid cleavage enzymes prevents HFD-induced inflammation with potential modulating gut microbiota and inhibits HFD-promoted HCC development.
Collapse
Affiliation(s)
- Hui Xia
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Chun Liu
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Cheng-Chung Li
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Maobin Fu
- Nature and Wellness Research Department, Research and Development Division, Kagome Co., Ltd., Tochigi, Japan
| | - Shingo Takahashi
- Nature and Wellness Research Department, Research and Development Division, Kagome Co., Ltd., Tochigi, Japan
| | - Kang-Quan Hu
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Koichi Aizawa
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts.,Nature and Wellness Research Department, Research and Development Division, Kagome Co., Ltd., Tochigi, Japan
| | - Suganuma Hiroyuki
- Nature and Wellness Research Department, Research and Development Division, Kagome Co., Ltd., Tochigi, Japan
| | - Guojun Wu
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey
| | - Liping Zhao
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey
| | - Xiang-Dong Wang
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts.
| |
Collapse
|
113
|
Jang IA, Kim EN, Lim JH, Kim MY, Ban TH, Yoon HE, Park CW, Chang YS, Choi BS. Effects of Resveratrol on the Renin-Angiotensin System in the Aging Kidney. Nutrients 2018; 10:E1741. [PMID: 30424556 PMCID: PMC6267480 DOI: 10.3390/nu10111741] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system (RAS), especially the angiotensin II (Ang II)/angiotensin II type 1 receptor (AT1R) axis, plays an important role in the aging process of the kidney, through increased tissue reactive oxygen species production and progressively increased oxidative stress. In contrast, the angiotensin 1-7 (Ang 1-7)/Mas receptor (MasR) axis, which counteracts the effects of Ang II, is protective for end-organ damage. To evaluate the ability of resveratrol (RSV) to modulate the RAS in aging kidneys, eighteen-month-old male C57BL/6 mice were divided into two groups that received either normal mouse chow or chow containing resveratrol, for six months. Renal expressions of RAS components, as well as pro- and antioxidant enzymes, were measured and mouse kidneys were isolated for histopathology. Resveratrol-treated mice demonstrated better renal function and reduced albuminuria, with improved renal histologic findings. Resveratrol suppressed the Ang II/AT1R axis and enhanced the AT2R/Ang 1-7/MasR axis. Additionally, the expression of nicotinamide adenine dinucleotide phosphate oxidase 4, 8-hydroxy-2'-deoxyguanosine, 3-nitrotyrosine, collagen IV, and fibronectin was decreased, while the expression of endothelial nitric oxide synthase and superoxide dismutase 2 was increased by resveratrol treatment. These findings demonstrate that resveratrol exerts protective effects on aging kidneys by reducing oxidative stress, inflammation, and fibrosis, through Ang II suppression and MasR activation.
Collapse
Affiliation(s)
- In-Ae Jang
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Eun Nim Kim
- Division of Medical Cell Biology, Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Ji Hee Lim
- Division of Medical Cell Biology, Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Min Young Kim
- Division of Medical Cell Biology, Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Tae Hyun Ban
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul 06591, Korea.
| | - Hye Eun Yoon
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary's Hospital, Incheon 21431, Korea.
| | - Cheol Whee Park
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul 06591, Korea.
| | - Yoon Sik Chang
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary's Hospital, Seoul 07345, Korea.
| | - Bum Soon Choi
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, St. Paul's Hospital, Seoul 02559, Korea.
| |
Collapse
|
114
|
Liang X, Liu Y, Jia S, Xu X, Dong M, Wei Y. SIRT1: The Value of Functional Outcome, Stroke-Related Dementia, Anxiety, and Depression in Patients with Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2018; 28:205-212. [PMID: 30361109 DOI: 10.1016/j.jstrokecerebrovasdis.2018.09.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/15/2018] [Accepted: 09/23/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The outcome of ischemic stroke depends on multiple factors and their function of each other. Studies have shown that Sirtuin1 (SIRT1) plays a chief role in the key procedure during ischemia/hypoxia by protecting against cellular stress and controlling the metabolic pathways. AIMS To explore the alterations in serum SIRT1 concentrations in acute ischemic stroke (AIS) patients and the relationship between SIRT1 and poststroke dementia, anxiety, and depression. METHODS One hundred and twenty four consecutive patients with clinically diagnosed AIS were recruited to participate in the study. Serum SIRT1 levels were measured using a commercially available ELISA equipment for SIRT1 (Cusabio, Wuhan, China). In 1 year after admission, the severity of stroke was assessed with the National Institutes of Health Stroke Scale score, and the functional outcome was measured by a modified Rankin scale, the Hamilton Anxiety Scale scores were evaluated to define patients with or without anxiety, and the Hamilton Depression Scale scores for depression. RESULTS We found the levels of serum SIRT1 was significantly higher (P = .036) in AIS patients (.62 ± .77 ng/mL) compared with healthy control subjects (.45 ± .69 ng/mL), but not significantly higher SIRT1 concentration (.58 ± .69 versus .64 ± .81 ng/mL, P = .298) than patients in the unfavorable functional outcome group. CONCLUSIONS There is no potential diagnostic and prognostic role of SIRT1 in AIS-related dementia, anxiety, and depression. The role of SIRT1 in AIS among human race needs to be further investigated.
Collapse
Affiliation(s)
- Xue Liang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China.
| | - Yang Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China.
| | - Shiyu Jia
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China.
| | - Xiaomin Xu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Meixue Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China.
| | - Youdong Wei
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China.
| |
Collapse
|
115
|
Lee Y, Kwon EY, Choi MS. Dietary Isoliquiritigenin at a Low Dose Ameliorates Insulin Resistance and NAFLD in Diet-Induced Obesity in C57BL/6J Mice. Int J Mol Sci 2018; 19:ijms19103281. [PMID: 30360437 PMCID: PMC6214092 DOI: 10.3390/ijms19103281] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/13/2018] [Accepted: 10/18/2018] [Indexed: 02/08/2023] Open
Abstract
Isoliquiritigenin (ILG) is a flavonoid constituent of Glycyrrhizae plants. The current study investigated the effects of ILG on diet-induced obesity and metabolic diseases. C57BL/6J mice were fed a normal diet (AIN-76 purified diet), high-fat diet (40 kcal% fat), and high-fat diet +0.02% (w/w) ILG for 16 weeks. Supplementation of ILG resulted in decreased body fat mass and plasma cholesterol level. ILG ameliorated hepatic steatosis by suppressing the expression of hepatic lipogenesis genes and hepatic triglyceride and fatty acid contents, while enhancing β-oxidation in the liver. ILG improved insulin resistance by lowering plasma glucose and insulin levels. This was also demonstrated by the intraperitoneal glucose tolerance test (IPGTT). Additionally, ILG upregulated the expression of insulin signaling-related genes in the liver and muscle. Interestingly, ILG elevated energy expenditure by increasing the expression of thermogenesis genes, which is linked to stimulated mitochondrial biogenesis and uncoupled cellular respiration in brown adipose tissue. ILG also suppressed proinflammatory cytokine levels in the plasma. These results suggest that ILG supplemented at 0.02% in the diet can ameliorate body fat mass, plasma cholesterol, non-alcoholic fatty liver disease, and insulin resistance; these effects were partly mediated by increasing energy expenditure in high-fat fed mice.
Collapse
Affiliation(s)
- Youngmi Lee
- Department of Food Science and Nutrition, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
| |
Collapse
|
116
|
Development of a high-content imaging assay for screening compound aggregation. Anal Biochem 2018; 559:30-33. [PMID: 30142329 DOI: 10.1016/j.ab.2018.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/01/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
Abstract
Aggregated compounds can promiscuously and nonspecifically associate with proteins resulting in either false inhibition or activation of many different protein target classes. We developed a high-content imaging assay in a 384-well format using fluorescently labeled target proteins and an Operetta cell imager to screen for compound aggregates that interact with target proteins. The high-throughput assay can not only directly detect the interaction between compound aggregators and the target of interest, but also determine the critical aggregation concentration (CAC) of a given promiscuous small molecule.
Collapse
|
117
|
Geng K, Fu N, Yang X, Xia W. Adjudin delays cellular senescence through Sirt3‑mediated attenuation of ROS production. Int J Mol Med 2018; 42:3522-3529. [DOI: 10.3892/ijmm.2018.3917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 01/06/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Keyi Geng
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Ningzhen Fu
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiao Yang
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
118
|
Zhang Y, Thai K, Jin T, Woo M, Gilbert RE. SIRT1 activation attenuates α cell hyperplasia, hyperglucagonaemia and hyperglycaemia in STZ-diabetic mice. Sci Rep 2018; 8:13972. [PMID: 30228292 PMCID: PMC6143559 DOI: 10.1038/s41598-018-32351-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
The NAD+-dependent lysine deacetylase, Sirtuin 1 (SIRT1), plays a central role in metabolic regulation. With type 1 diabetes a disease that is characterised by metabolic dysregulation, we sought to assess the impact of SIRT1 activation in experimental, streptozotocin (STZ)-induced diabetes. CD1 mice with and without STZ-induced diabetes were randomized to receive the SIRT1 activating compound, SRT3025, or vehicle over 20 weeks. Vehicle treated STZ-CD1 mice developed severe hyperglycaemia with near-absent circulating insulin and widespread beta cell loss in association with hyperglucagonaemia and expanded islet alpha cell mass. Without affecting ß-cell mass or circulating insulin, diabetic mice that received SRT3025 had substantially improved glycaemic control with greatly reduced islet α cell mass and lower plasma glucagon concentrations. Consistent with reduced glucagon abundance, the diabetes-associated overexpression of key gluconeogenic enzymes, glucose-6-phosphatase and PEPCK were also lowered by SRT3025. Incubating cultured α cells with SRT3025 diminished their glucagon secretion and proliferative activity in association with a reduction in the α cell associated transcription factor, Aristaless Related Homeobox (Arx). By reducing the paradoxical increase in glucagon, SIRT1 activation may offer a new, α-cell centric approach to the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Yanling Zhang
- St. Michael's Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, M5B 1W8, Canada
| | - Kerri Thai
- St. Michael's Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, M5B 1W8, Canada
| | - Tianru Jin
- Toronto General Hospital Research Institute (TGHRI), Toronto, ON, M5G 2C4, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute (TGHRI), Toronto, ON, M5G 2C4, Canada
| | - Richard E Gilbert
- St. Michael's Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, M5B 1W8, Canada.
| |
Collapse
|
119
|
Zhang L, Wei C, Ruan Y, Zhang Y, Zhou Y, Lei D. Serum containing Buyang Huanwu decoction prevents age-associated migration and invasion of human vascular smooth muscle cells by up regulating SIRT1 expression. Biosci Trends 2018; 12:282-290. [PMID: 29952352 DOI: 10.5582/bst.2018.01063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The migration and invasion of vascular smooth muscle cells (VSMCs) caused by advanced aging play an important role in diffuse intimal thickening, facilitate adverse arterial remodeling and contribute to the initiation and progression of cardiovascular diseases. The inhibitory function of Buyang Huanwu decoction (BYHWD) has been found on aortic intimal hyperplasia and VSMC proliferation, but its effect on age-associated migration and invasion remains unknown. Here, we used an in vitro angiotensin II (Ang II)-induced senescence model to study the effects of serum containing BYHWD (BYHWS) on the migratory and invasive capacities, matrix metalloprotease type 2 (MMP-2) expression and modulation of sirtuin1 (SIRT1) signaling in human aorta VSMCs (HA-VAMCs). Our results showed that BYHWS was able to inhibit Ang II-induced migration and invasion, with down-regulation of MMP-2. In addition, manipulation of SIRT1 by either over-expression or siRNA knockdown ameliorated or promoted cellular migration and invasion, respectively. Moreover, BYHWS reversed senescence-mediated decrease of SIRT1 levels and SIRT1 was required for BYHWS regulation on migration and invasion of senescent HA-VAMCs. In summary, our data demonstrated that BYHWS suppressed the migration and invasion of age-associated VSMC via an increase of the SIRT1 level, which provides novel insights for the therapy of age-associated cardiovascular diseases.
Collapse
MESH Headings
- Aging/drug effects
- Aging/physiology
- Angiotensin II/pharmacology
- Aorta/cytology
- Aorta/physiology
- Cardiovascular Diseases/drug therapy
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cellular Senescence/drug effects
- Cellular Senescence/physiology
- Down-Regulation
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Gene Knockdown Techniques
- Humans
- Matrix Metalloproteinase 2/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- RNA, Small Interfering/metabolism
- Signal Transduction/drug effects
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| | - Chunshan Wei
- Department of Liver Disease, Shenzhen Hospital Affiliated to Guangzhou University of Chinese Medicine
| | - Yunjun Ruan
- Department of Cardiology, Guangzhou General Hospital of Guangzhou Military Command
| | - Yanan Zhang
- Veterinary medicine, Northeast Agricultural University
| | - Yuliang Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| | - Da Lei
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| |
Collapse
|
120
|
Marcondes JPDC, Andrade PFB, Sávio ALV, Silveira MAD, Rudge MVC, Salvadori DMF. BCL2 and miR-181a transcriptional alterations in umbilical-cord blood cells can be putative biomarkers for obesity. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 836:90-96. [PMID: 30442352 DOI: 10.1016/j.mrgentox.2018.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/17/2018] [Accepted: 06/01/2018] [Indexed: 10/14/2022]
Abstract
Several findings suggest that in utero stressor stimuli can alter fetal development by promoting transcriptional changes, and predisposing the neonate to diseases later in life. This study aimed to investigate whether a hyperglycemic environment in pregnant women with gestational diabetes mellitus (GDM) is able to cause fetal genetic alterations and predispose neonates to obesity. Transcriptional alteration of SIRT1, TP53 and BCL2 genes, miR-181a (a SIRT1 or BCL2 regulator) and telomere length were evaluated in placental and umbilical-cord blood cells. Healthy (HP; n = 20) and GDM (n = 20) pregnant women and their respective neonates were included in the study. Additionally, obese (n = 20) and eutrophic (n = 20) adults also participated as reference populations. Gene expression data showed down-regulation of BCL2 in umbilical-cord and peripheral blood cells from GDM neonates and obese adults, respectively. The miR-181a was down-regulated only in umbilical-cord blood cells of GDM neonates. Telomere length presented no significant difference. In conclusion, our study demonstrated that the GDM hyperglycemic intrauterine environment promotes transcriptional alterations in BCL2 and miR-181a in neonate umbilical-cord blood cells. Furthermore, both GDM neonates and obese subjects share the same transcriptional alteration in BCL2. Considering the relationship between obesity development and the functions regulated by these two genes, BCL2 and miR-181a could be adopted as potential biomarkers for childhood obesity. However, further study designs are recommended to confirm this hypothesis.
Collapse
Affiliation(s)
- João Paulo de Castro Marcondes
- UNESP - São Paulo State University, Medical School, Botucatu, SP, Brazil; UNESP - São Paulo State University, Bioscience Institute, Botucatu, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
121
|
Sun X, Wang P, Yao LP, Wang W, Gao YM, Zhang J, Fu YJ. Paeonol alleviated acute alcohol-induced liver injury via SIRT1/Nrf2/NF-κB signaling pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:110-117. [PMID: 29704732 DOI: 10.1016/j.etap.2018.04.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 05/22/2023]
Abstract
In this study, the beneficial effect of paeonol on acute alcohol-induced liver injury and the basic mechanisms were investigated. in vitro, HepG2 cells were treated with paeonol for 24 h before it were exposed to alcohol for 24 h. in vivo, male C57BL/6 mice were used to establish alcohol-induced liver injury models by oral gavage of alcohol (5 g/kg BW). Paeonol pretreatment showed statistically significant reduction in alcohol-induced ROS, MDA, IL-1β, IL-6, TNF-α, and nitric oxide, while GSH content was retained (P < 0.05). Furthermore, paeonol treatment resulted in the increase of Nrf2 nuclear translocation, the increase of NQO-1 and HO-1 expression, and the suppression of NF-κB p65 nuclear translocation. However, pretreatment with NAM (inhibitor of SIRT1) not only inhibited the effect of paeonol on reducing nuclear translocation of NF-κBp65, but also inhibited the effect of paeonol on promoting the expression of nuclear Nrf2, NQO1 and HO-1. Besides, paeonol pretreatment at test doses significantly ameliorated alcohol-induced edema, hepatocyte necrosis and hepatic cord irregular. These results demonstrated that paeonol has the high potential for relieving acute alcohol-induced liver injury.
Collapse
Affiliation(s)
- Xing Sun
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Peng Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Li-Ping Yao
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Wei Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Yi-Meng Gao
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Jing Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Yu-Jie Fu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Harbin 150040, PR China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China; The college of Forestry, Beijing Forestry University, Beijing 100083, PR China.
| |
Collapse
|
122
|
SIRT1 induces resistance to apoptosis in human granulosa cells by activating the ERK pathway and inhibiting NF-κB signaling with anti-inflammatory functions. Apoptosis 2018; 22:1260-1272. [PMID: 28755171 DOI: 10.1007/s10495-017-1386-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIRT1, a member of the sirtuin family, has recently emerged as a vital molecule in controlling ovarian function. The aims of the present study were to investigate SIRT1 expression and analyze SIRT1-mediated apoptosis in human granulosa cells (GCs). Human ovarian tissues were subjected to immunohistochemistry for localization of SIRT1 expression. SIRT1 knockdown in a human ovarian GC tumor line (COV434) was achieved by small interfering RNA, and the relationship between apoptosis and SIRT1 was assessed by quantitative reverse transcription polymerase chain reaction and western blotting. We further detected SIRT1 expression in human luteinized GCs. Associations among SIRT1 knockdown, SIRT1 stimulation (resveratrol) and expression of ERK1/2 and apoptotic regulatory proteins were analyzed in cell lines and luteinized GCs. Resveratrol downregulated the levels of nuclear factor (NF)-κB/p65, but this inhibitory effect was attenuated by suppressing SIRT1 activity. The NF-κB/p65 inhibitor pyrrolidine dithiocarbamate achieved similar anti-apoptosis effects. These results suggest that SIRT1 might play an anti-apoptotic role in apoptosis processes in GCs, possibly by sensing and regulating the ERK1/2 pathway, which has important clinical implications. Thus, our study provides a mechanistic link, whereby activation of SIRT1 function might help to sustain human reproduction by maintaining GCs as well as oocytes, offering a novel approach for developing a new class of therapeutic anti-inflammatory agents.
Collapse
|
123
|
Luo FQ, Ma Q, Zheng H, Guo XW, Zhang J. Involvement of spinal SIRT1 in development of chronic constriction injury induced neuropathic pain in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:2561-2569. [PMID: 31938369 PMCID: PMC6958300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/27/2016] [Indexed: 06/10/2023]
Abstract
It is known that the epigenetic process of histone acetylation is involved in the neuropathic pain. The aim of this study was to determine whether sirtuin type 1 (SIRT1), an NAD+ dependent deacetylase, affected allodynia and hyperalgesia in neuropathic pain. The neuropathic pain model was established by ligature of the right sciatic nerve to induce chronic constriction injury (CCI) in rats. Histone acetyltransferase (HAT) activity was increased and, and histone deacetylase (HDAC) activity was declined in tissue of the spinal dorsa horn in CCI rates by means of enzyme-linked immunosorbent assay (ELISA). The persistent hyperalgesia and allodynia caused by CCI were associated with downregulation of SIRT1 and upregulation of acetylated-H3 (Ac-H3) in tissue of the spinal cord by Western blot assay, which was reversed after intrathecal injection of SIRT1 agonist SRT1720. SRT1720 treatment achieved analgesic through inhibiting the acetylation of nuclear factor kappa B (NF-κB) and blocking the releases of the inflammatory factors including tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 by means of Western blot and real-time quantitative PCR (RT-PCR), respectively. Taken together, these data suggest that SIRT1 in the spinal cord plays an important role in the neuropathic pain in the rat model.
Collapse
Affiliation(s)
- Feng-Qin Luo
- Department of Anesthesiology, Zhejiang Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Traditional Chinese Medical University Hangzhou, Zhejiang Provincial, China
| | - Qian Ma
- Department of Anesthesiology, Zhejiang Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Traditional Chinese Medical University Hangzhou, Zhejiang Provincial, China
| | - Hui Zheng
- Department of Anesthesiology, Zhejiang Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Traditional Chinese Medical University Hangzhou, Zhejiang Provincial, China
| | - Xiao-Wen Guo
- Department of Anesthesiology, Zhejiang Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Traditional Chinese Medical University Hangzhou, Zhejiang Provincial, China
| | - Juan Zhang
- Department of Anesthesiology, Zhejiang Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Traditional Chinese Medical University Hangzhou, Zhejiang Provincial, China
| |
Collapse
|
124
|
Piperlongumine activates Sirtuin1 and improves cognitive function in a murine model of Alzheimer’s disease. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
125
|
Pan X, Chen J, Wang W, Chen L, Wang L, Ma Q, Zhang J, Chen L, Wang G, Zhang M, Wu H, Cheng R. Resveratrol-induced antinociception is involved in calcium channels and calcium/caffeine-sensitive pools. Oncotarget 2018; 8:9399-9409. [PMID: 28030799 PMCID: PMC5354740 DOI: 10.18632/oncotarget.14090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/15/2016] [Indexed: 11/25/2022] Open
Abstract
Resveratrol has been widely investigated for its potential health properties, although little is known about its mechanism in vivo. Previous studies have indicated that resveratrol produces antinociceptive effects in mice. Calcium channels and calcium/caffeine-sensitive pools are reported to be associated with analgesic effect. The present study was to explore the involvement of Ca2+ channel and calcium/caffeine-sensitive pools in the antinociceptive response of resveratrol. Tail-flick test was used to assess antinociception in mice treated with resveratrol or the combinations of resveratrol with MK 801, nimodipine, CaCl2, ryanodine and ethylene glycol tetraacetic acid (EGTA), respectively. The Ca2+/calmodulin-dependent protein kinase II (CaMKII) and brain-derived neurotrophic factor (BDNF) levels in the spinal cord were also investigated when treated with the above drugs. The results showed that resveratrol increased the tail flick latency in the tail-flick test, in dose-dependent manner. N-methyl-D-aspartate (NMDA) glutamate receptor antagonist MK 801 potentiated the antinociceptive effects of sub-threshold dose of resveratrol at 10 mg/kg. Ca2+ channel blocker, however, abolished the antinociceptive effects of resveratrol. In contrast to these results, EGTA or ryanodine treatment (i.c.v.) potentiated resveratrol-induced antinociception. There was a significant decrease in p-CaMKII and an increase in BDNF expression in the spinal cord when combined with MK 801, nimodipine, ryanodine and EGTA. While an increase in p-CaMKII level and a decrease in BDNF expression were observed when high dose of resveratrol combined with CaCl2. These findings suggest that resveratrol exhibits the antinociceptive effects by inhibition of calcium channels and calcium/caffeine-sensitive pools.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Department of Thyroid Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jiechun Chen
- Department of Neurology, Lianyungang second people's Hospital, Lianyungang, Jiangsu Province, China
| | - Weijie Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Ling Chen
- Department of Clinical Pharmacy, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Lin Wang
- Department of Clinical Pharmacy, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Quan Ma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY
| | - Jianbo Zhang
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Lichao Chen
- Pingyang Hospital of Traditional Chinese Medicine, Pingyang, Zhejiang Province, China
| | - Gang Wang
- Department of Clinical Pharmacy, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Meixi Zhang
- Pingyang Hospital of Traditional Chinese Medicine, Pingyang, Zhejiang Province, China
| | - Hao Wu
- Department of Neurology, Wenzhou People's Hospital, Wenzhou, Zhejiang Province, China
| | - Ruochuan Cheng
- Department of Thyroid Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
126
|
Qiu D, Hou X, Han L, Li X, Ge J, Wang Q. Sirt2-BubR1 acetylation pathway mediates the effects of advanced maternal age on oocyte quality. Aging Cell 2018; 17:e12698. [PMID: 29067790 PMCID: PMC5770883 DOI: 10.1111/acel.12698] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2017] [Indexed: 12/27/2022] Open
Abstract
The level of Sirt2 protein is reduced in oocytes from aged mice, while exogenous expression of Sirt2 could ameliorate the maternal age-associated meiotic defects. To date, the underlying mechanism remains unclear. Here, we confirmed that specific depletion of Sirt2 disrupts maturational progression and spindle/chromosome organization in mouse oocytes, with compromised kinetochore-microtubule attachments. Candidate screening revealed that acetylation state of lysine 243 on BubR1 (BubR1-K243, an integral part of the spindle assembly checkpoint complex) functions during oocyte meiosis, and acetylation-mimetic mutant BubR1-K243Q results in the very similar phenotypes as Sirt2-knockdown oocytes. Furthermore, we found that nonacetylatable-mimetic mutant BubR1-K243R partly prevents the meiotic deficits in oocytes depleted of Sirt2. Importantly, BubR1-K243R overexpression in oocytes derived from aged mice markedly suppresses spindle/chromosome anomalies and thereupon lowers the incidence of aneuploid eggs. In sum, our data suggest that Sirt2-dependent BubR1 deacetylation involves in the regulation of meiotic apparatus in normal oocytes and mediates the effects of advanced maternal age on oocyte quality.
Collapse
Affiliation(s)
- Danhong Qiu
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Xiaojing Hou
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Longsen Han
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Xiaoyan Li
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
- College of Animal Science & TechnologyNanjing Agricultural UniversityNanjingChina
| | - Juan Ge
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Qiang Wang
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|
127
|
Wong SY, Tang BL. SIRT1 as a therapeutic target for Alzheimer's disease. Rev Neurosci 2018; 27:813-825. [PMID: 27497424 DOI: 10.1515/revneuro-2016-0023] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/12/2016] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia in the aging population worldwide. SIRT1 deacetylation of histones and transcription factors impinge on multiple neuronal and non-neuronal targets, and modulates stress response, energy metabolism and cellular senescence/death pathways. Collectively, SIRT1 activity could potentially affect multiple aspects of hippocampal and cortical neuron function and survival, thus modifying disease onset and progression. In this review, the known and potential mechanisms of action of SIRT1 with regard to AD, and its potential as a therapeutic target, are discussed.
Collapse
|
128
|
Yamamoto M, Takahashi Y. The Essential Role of SIRT1 in Hypothalamic-Pituitary Axis. Front Endocrinol (Lausanne) 2018; 9:605. [PMID: 30405528 PMCID: PMC6205959 DOI: 10.3389/fendo.2018.00605] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/24/2018] [Indexed: 01/28/2023] Open
Abstract
The endocrine system plays an essential role in the physiological adaptation to malnutrition. The adaptive response of various hormones directs the energy utilization toward the survival functions and away from growth and reproduction. Particularly, the hypothalamic pituitary axis plays an integral and a central role in the regulation of endocrine organs. Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase that is activated in response to calorie restriction (CR). SIRT1 is involved in cellular processes via the deacetylation of histone as well as various transcription factors and signal transduction molecules and thereby modulates the endocrine/metabolic functions. There is much evidence to demonstrate clearly that SIRT1 in the hypothalamus, pituitary gland, and other target organs modifies the synthesis, secretion, and activities of hormones and in turn induces the adaptive responses. In this review, we discussed the role of SIRT1 in the hypothalamic pituitary axis and its pathophysiological significance.
Collapse
Affiliation(s)
- Masaaki Yamamoto
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- *Correspondence: Yutaka Takahashi
| |
Collapse
|
129
|
Truong VL, Jun M, Jeong WS. Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors 2018; 44:36-49. [PMID: 29193412 DOI: 10.1002/biof.1399] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022]
Abstract
Resveratrol, a natural polyphenolic compound, is found in various kinds of fruits, plants, and their commercial products such as red wine. It has been demonstrated to exhibit a variety of health-promoting effects including prevention and/or treatment of cardiovascular diseases, inflammation, diabetes, neurodegeneration, aging, and cancer. Cellular defensive properties of resveratrol can be explained through its ability of either directly neutralizing reactive oxygen species/reactive nitrogen species (ROS/RNS) or indirectly upregulating the expression of cellular defensive genes. As a direct antioxidant agent, resveratrol scavenges diverse ROS/RNS as well as secondary organic radicals with mechanisms of hydrogen atom transfer and sequential proton loss electron transfer, thereby protecting cellular biomolecules from oxidative damage. Resveratrol also enhances the expression of various antioxidant defensive enzymes such as heme oxygenase 1, catalase, glutathione peroxidase, and superoxide dismutase as well as the induction of glutathione level responsible for maintaining the cellular redox balance. Such defenses could be achieved by regulating various signaling pathways including sirtuin 1, nuclear factor-erythroid 2-related factor 2 and nuclear factor κB. This review provides current understanding and information on the role of resveratrol in cellular defense system against oxidative stress. © 2017 BioFactors, 44(1):36-49, 2018.
Collapse
Affiliation(s)
- Van-Long Truong
- Department of Food and Life Sciences, College of BNIT, Inje University, Gimhae 50834, Korea
| | - Mira Jun
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Korea
| | - Woo-Sik Jeong
- Department of Food and Life Sciences, College of BNIT, Inje University, Gimhae 50834, Korea
| |
Collapse
|
130
|
Liao M, Wang J. Mechanisms of Hematopoietic Stem Cell Ageing and Targets for Hematopoietic Tumour Prevention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1086:117-140. [PMID: 30232756 DOI: 10.1007/978-981-13-1117-8_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hematopoietic stem cells represent a rare population in the bone marrow, with the capacity of generating all blood lineage and themselves at the same time. With aging, the reconstitution capacity of hematopoietic stem cells decreases accompanying with differentiation skewing wherein the myeloid branch dominates in both mouse and human. In recent years, various molecular mechanisms that induce functional decline of HSC during aging were disclosed including DNA damage accumulation, metabolic alteration, defects in protein homeostasis, and aging-induced changes in the blood circulatory environment. Deciphering the nature of HSC aging could improve our knowledge of HSC aging-related diseases and furthermore promote the developing of therapeutic interventions for human HSC aging and diseases.
Collapse
Affiliation(s)
- Min Liao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
131
|
Abe J, Yamada Y, Takeda A, Harashima H. Cardiac progenitor cells activated by mitochondrial delivery of resveratrol enhance the survival of a doxorubicin-induced cardiomyopathy mouse model via the mitochondrial activation of a damaged myocardium. J Control Release 2018; 269:177-188. [DOI: 10.1016/j.jconrel.2017.11.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/24/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
|
132
|
Sirtuins as Modifiers of Huntington's Disease (HD) Pathology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 154:105-145. [DOI: 10.1016/bs.pmbts.2017.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
133
|
Emerging Roles of Sirtuin 6 in Alzheimer's Disease. J Mol Neurosci 2017; 64:157-161. [PMID: 29260452 DOI: 10.1007/s12031-017-1005-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that is imposing an increasing burden on society. Currently, AD is the leading cause of senile dementia worldwide. Despite the long existence of AD, there is lack of therapies for AD, suggesting that new and effective treatment strategy must be explored. At present, sirtuin pathway has attracted attention from the researchers due to its promising results in laboratory models of aging. In addition, our understanding in the roles of sirtuin 6 in AD has expanded. It has been identified to be involved in telomere maintenance, DNA repair, genome integrity, energy metabolism, and inflammation, which ultimately regulate life span. Recent findings also demonstrate that sirtuin 6 is lacking in AD patients, proposing that it can be a new potential therapeutic target in AD. Therefore, exploring on how sirtuin 6 is related in AD manifestation may accelerate the research of AD further and benefits future AD patients. Keeping that in mind, this review aims to highlight the possible roles of sirtuin 6 in AD manifestation.
Collapse
|
134
|
Pollack RM, Barzilai N, Anghel V, Kulkarni AS, Golden A, O'Broin P, Sinclair DA, Bonkowski MS, Coleville AJ, Powell D, Kim S, Moaddel R, Stein D, Zhang K, Hawkins M, Crandall JP. Resveratrol Improves Vascular Function and Mitochondrial Number but Not Glucose Metabolism in Older Adults. J Gerontol A Biol Sci Med Sci 2017; 72:1703-1709. [PMID: 28329397 DOI: 10.1093/gerona/glx041] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/27/2017] [Indexed: 02/04/2023] Open
Abstract
Background Resveratrol, a plant-derived polyphenol, has been reported to improve glucose metabolism and vascular function and to extend life span in animal models, but studies in humans have been inconclusive. Methods In a randomized, double-blind crossover study, we treated older glucose-intolerant adults (n = 30) with resveratrol (2-3 g/daily) or placebo, each for 6 weeks. A standard mixed-meal test was used to assess insulin sensitivity (Matsuda index) and secretion (C-peptide deconvolution) and vascular function by reactive hyperemia peripheral arterial tonometry. Skeletal muscle samples were obtained for gene expression using RNA-Seq analysis and to assess mitochondrial morphology. Results There were no changes in glucose tolerance, insulin sensitivity, weight, blood pressure, or lipid profile following resveratrol treatment. Fasting reactive hyperemia index improved with resveratrol (2.02 ± 0.2 vs 1.76 ± 0.02, p = .002). RNA-Seq analysis yielded 140 differentially expressed transcripts (corrected p-value ≤ .05), predominantly associated with mitochondrial genes and noncoding RNA. Ingenuity Pathway Analysis confirmed that mitochondrial dysfunction (p = 2.77 × 10-12) and oxidative phosphorylation (p = 1.41 × 10-11) were the most significantly perturbed pathways. Mitochondrial number, but not size, was increased. Conclusions Resveratrol treatment of older adults with impaired glucose regulation may have beneficial effects on vascular function, but not glucose metabolism or insulin sensitivity. Changes in gene expression suggest effects similar to those observed with caloric restriction, which has been shown to increase life and health span in animal models, although its significance for humans is uncertain. Future human studies should address the appropriate dose range and low bioavailability of resveratrol.
Collapse
Affiliation(s)
| | - Nir Barzilai
- Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Aaron Golden
- Albert Einstein College of Medicine, Bronx, New York
| | - Pilib O'Broin
- National University of Ireland Galway, Galway, Ireland
| | | | | | | | | | - Sharon Kim
- Albert Einstein College of Medicine, Bronx, New York
| | - Ruin Moaddel
- National Institute on Aging, Baltimore, Maryland
| | - Daniel Stein
- Albert Einstein College of Medicine, Bronx, New York
| | - Kehao Zhang
- Albert Einstein College of Medicine, Bronx, New York
| | | | | |
Collapse
|
135
|
Al-Hussaini H, Kilarkaje N. Trans-resveratrol mitigates type 1 diabetes-induced oxidative DNA damage and accumulation of advanced glycation end products in glomeruli and tubules of rat kidneys. Toxicol Appl Pharmacol 2017; 339:97-109. [PMID: 29229234 DOI: 10.1016/j.taap.2017.11.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022]
Abstract
Hyperglycemia induces the formation of advanced glycation end products (AGEs) and their receptors (RAGEs), which alter several intracellular signaling mechanisms leading to the onset and progression of diabetic nephropathy. The present study focused on, i) modulatory effects of trans-resveratrol (3,5,4'-trihydroxy-trans-stilbene) on structural changes, AGE (NƐ-carboxymethyl-lysine), RAGE, oxidative stress and DNA damage, and apoptosis, and ii) localization of fibrotic changes, AGE, RAGE, 8-oxo-dG and 4-hydroxynonenal (4-HNE) in diabetic rat kidneys. Resveratrol (5mg/kg; po, administered during last 45days of 90-day-long hyperglycemic period) administration to streptozotocin-induced type 1 diabetic male Wistar rats reduced renal hypertrophy and structural changes (tubular atrophy, mesangial expansion or shrinkage, diffuse glomerulonephritis, and fibrosis), AGE accumulation, oxidative stress and DNA damage (8-oxo-dG), 4-HNE, caspase-3, and cleaved-caspase-3, but not the RAGE expression. The AGE accumulated in the mesangium, vascular endothelium, and proximal convoluted tubules and less intensely in distal convoluted tubules of diabetic rat kidneys. The RAGE expression increased in the convoluted tubules and collecting ducts of diabetic rat kidneys, but not in the mesangium. Diabetes increased the expression of 8-oxo-dG in nuclei and cytoplasm of renal cells, and 4-HNE in glomeruli, convoluted tubules, the loops of Henle and collecting ducts. Hyperglycemia-induced AGE-RAGE axis and oxidative stress in turn induced apoptosis in diabetic kidneys. Resveratrol mitigated all diabetic effects except the RAGE expression. In conclusion, Resveratrol significantly alleviates diabetes-induced glycation, oxidative damage, and apoptosis to inhibit the progression of diabetic nephropathy. Resveratrol supplementation may be useful to hinder the onset and progression of diabetic kidney diseases.
Collapse
Affiliation(s)
- Heba Al-Hussaini
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | | |
Collapse
|
136
|
Saihara K, Kamikubo R, Ikemoto K, Uchida K, Akagawa M. Pyrroloquinoline Quinone, a Redox-Active o-Quinone, Stimulates Mitochondrial Biogenesis by Activating the SIRT1/PGC-1α Signaling Pathway. Biochemistry 2017; 56:6615-6625. [DOI: 10.1021/acs.biochem.7b01185] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Kazuhiro Saihara
- Department
of Biological Chemistry, Division of Applied Life Science, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 599-8531, Japan
| | - Ryosuke Kamikubo
- Department
of Biological Chemistry, Division of Applied Life Science, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 599-8531, Japan
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Kazuto Ikemoto
- Niigata
Research Laboratory, Mitsubishi Gas Chemical Company, Inc., Niigata 950-3112, Japan
| | - Koji Uchida
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Mitsugu Akagawa
- Department
of Biological Chemistry, Division of Applied Life Science, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 599-8531, Japan
| |
Collapse
|
137
|
Yu J, Qin B, Wu F, Qin S, Nowsheen S, Shan S, Zayas J, Pei H, Lou Z, Wang L. Regulation of Serine-Threonine Kinase Akt Activation by NAD +-Dependent Deacetylase SIRT7. Cell Rep 2017; 18:1229-1240. [PMID: 28147277 DOI: 10.1016/j.celrep.2017.01.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/02/2016] [Accepted: 01/06/2017] [Indexed: 12/15/2022] Open
Abstract
The Akt pathway is a central regulator that promotes cell survival in response to extracellular signals. Depletion of SIRT7, an NAD+-dependent deacetylase that is the least-studied sirtuin, is known to significantly increase Akt activity in mice through unknown mechanisms. In this study, we demonstrate that SIRT7 depletion in breast cancer cells results in Akt hyper-phosphorylation and increases cell survival following genotoxic stress. Mechanistically, SIRT7 specifically interacts with and deacetylates FKBP51 at residue lysines 28 and 155 (K28 and K155), resulting in enhanced interactions among FKBP51, Akt, and PHLPP, as well as Akt dephosphorylation. Mutating both lysines to arginines abolishes the effect of SIRT7 on Akt activity through FKBP51 deacetylation. Finally, energy stress strengthens SIRT7-mediated effects on Akt dephosphorylation through FKBP51 and thus sensitizes cancer cells to cytotoxic agents. These results reveal a direct role of SIRT7 in Akt regulation and raise the possibility of using the glucose analog 2-deoxy-D-glucose (2DG) as a chemo-sensitizing agent.
Collapse
Affiliation(s)
- Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Bo Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Fengying Wu
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Sisi Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Somaira Nowsheen
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic School of Medicine and the Mayo Clinic Medical Scientist Training Program, Rochester, MN 55905, USA
| | - Shan Shan
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jacqueline Zayas
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic School of Medicine and the Mayo Clinic Medical Scientist Training Program, Rochester, MN 55905, USA
| | - Huadong Pei
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA; Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhenkun Lou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
138
|
Sirt6 Promotes DNA End Joining in iPSCs Derived from Old Mice. Cell Rep 2017; 18:2880-2892. [PMID: 28329681 DOI: 10.1016/j.celrep.2017.02.082] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/22/2017] [Accepted: 02/28/2017] [Indexed: 12/11/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have great potential for treating age-related diseases, but the genome integrity of iPSCs is critically important. Here, we demonstrate that non-homologous end joining (NHEJ), rather than homologous recombination (HR), is less efficient in iPSCs from old mice than young mice. We further find that Sirt6 is downregulated in iPSCs from old mice. Sirt6 directly binds to Ku80 and facilitates the Ku80/DNA-PKcs interaction, thus promoting DNA-PKcs phosphorylation at residue S2056, leading to efficient NHEJ. Rescue experiments show that introducing a combination of Sirt6 and the Yamanaka factors during reprogramming significantly promotes DNA double-strand break (DSB) repair by activating NHEJ in iPSCs derived from old mice. Thus, our study suggests a strategy to improve the quality of iPSCs derived from old donors by activating NHEJ and stabilizing the genome.
Collapse
|
139
|
Xu S, Yin M, Koroleva M, Mastrangelo MA, Zhang W, Bai P, Little PJ, Jin ZG. SIRT6 protects against endothelial dysfunction and atherosclerosis in mice. Aging (Albany NY) 2017; 8:1064-82. [PMID: 27249230 PMCID: PMC4931854 DOI: 10.18632/aging.100975] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/18/2016] [Indexed: 12/31/2022]
Abstract
SIRT6 is an important member of sirtuin family that represses inflammation, aging and DNA damage, three of which are causing factors for endothelial dysfunction. SIRT6 expression is decreased in atherosclerotic lesions from ApoE−/− mice and human patients. However, the role of SIRT6 in regulating vascular endothelial function and atherosclerosis is not well understood. Here we show that SIRT6 protects against endothelial dysfunction and atherosclerosis. Global and endothelium-specific SIRT6 knockout mice exhibited impaired endothelium-dependent vasorelaxation. Moreover, SIRT6+/− haploinsufficient mice fed a high-fat diet (HFD) also displayed impaired endothelium-dependent vasorelaxation. Importantly, SIRT6+/−;ApoE−/− mice after HFD feeding exhibited exacerbated atherosclerotic lesion development, concurrent with increased expression of the proinflammatory cytokine VCAM-1. Loss- and gain-of-SIRT6 function studies in cultured human endothelial cells (ECs) showed that SIRT6 attenuated monocyte adhesion to ECs. RNA-sequencing profiling revealed that SIRT6 overexpression decreased the expression of multiple atherosclerosis-related genes, including proatherogenic gene TNFSF4 (tumor necrosis factor superfamily member 4). Chromatin immunoprecipitation assays showed that SIRT6 decreased TNFSF4 gene expression by binding to and deacetylating H3K9 at TNFSF4 gene promoter. Collectively, these findings demonstrate that SIRT6 play a pivotal role in maintaining endothelial function and increased SIRT6 activity could be a new therapeutic strategy to combat atherosclerotic disease.
Collapse
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| | - Meimei Yin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| | - Marina Koroleva
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| | - Michael A Mastrangelo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, 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
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence (PACE), Woolloongabba QLD 4102, Australia
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| |
Collapse
|
140
|
Pannek M, Simic Z, Fuszard M, Meleshin M, Rotili D, Mai A, Schutkowski M, Steegborn C. Crystal structures of the mitochondrial deacylase Sirtuin 4 reveal isoform-specific acyl recognition and regulation features. Nat Commun 2017; 8:1513. [PMID: 29138502 PMCID: PMC5686155 DOI: 10.1038/s41467-017-01701-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 10/07/2017] [Indexed: 12/22/2022] Open
Abstract
Sirtuins are evolutionary conserved NAD+-dependent protein lysine deacylases. The seven human isoforms, Sirt1-7, regulate metabolism and stress responses and are considered therapeutic targets for aging-related diseases. Sirt4 locates to mitochondria and regulates fatty acid metabolism and apoptosis. In contrast to the mitochondrial deacetylase Sirt3 and desuccinylase Sirt5, no prominent deacylase activity and structural information are available for Sirt4. Here we describe acyl substrates and crystal structures for Sirt4. The enzyme shows isoform-specific acyl selectivity, with significant activity against hydroxymethylglutarylation. Crystal structures of Sirt4 from Xenopus tropicalis reveal a particular acyl binding site with an additional access channel, rationalizing its activities. The structures further identify a conserved, isoform-specific Sirt4 loop that folds into the active site to potentially regulate catalysis. Using these results, we further establish efficient Sirt4 activity assays, an unusual Sirt4 regulation by NADH, and Sirt4 effects of pharmacological modulators.
Collapse
Affiliation(s)
- Martin Pannek
- Department of Biochemistry, University of Bayreuth, 95440, Bayreuth, Germany
| | - Zeljko Simic
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06108, Halle, Germany
| | - Matthew Fuszard
- Department of Biochemistry, University of Bayreuth, 95440, Bayreuth, Germany
| | - Marat Meleshin
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06108, Halle, Germany
| | - Dante Rotili
- Department of Chemistry and Technologies of Drugs, Pasteur Institute Italy, Cenci-Bolognetti Foundation, Sapienza University of Rome, 00185, Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Pasteur Institute Italy, Cenci-Bolognetti Foundation, Sapienza University of Rome, 00185, Rome, Italy
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06108, Halle, Germany
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, 95440, Bayreuth, Germany.
| |
Collapse
|
141
|
Arctigenin attenuates ischemic stroke via SIRT1-dependent inhibition of NLRP3 inflammasome. Biochem Biophys Res Commun 2017; 493:821-826. [DOI: 10.1016/j.bbrc.2017.08.062] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 08/17/2017] [Indexed: 01/12/2023]
|
142
|
Wang CY, Sun ZN, Wang MX, Zhang C. SIRT1 mediates salidroside-elicited protective effects against MPP + -induced apoptosis and oxidative stress in SH-SY5Y cells: involvement in suppressing MAPK pathways. Cell Biol Int 2017; 42:84-94. [PMID: 28851138 DOI: 10.1002/cbin.10864] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease, leading to tremor, rigidity, bradykinesia, and gait impairment. Salidroside has been reported to exhibit antioxidative and neuroprotective properties in PD. However, the underlying neuroprotective mechanisms effects of salidroside are poorly understood. Recently, a growing body of evidences suggest that silent information regulator 1 (SIRT1) plays important roles in the pathophysiology of PD. Hence, the present study investigated the roles of SIRT1 in neuroprotective effect of salidroside against N-methyl-4-phenylpyridinium (MPP+ )-induced SH-SY5Y cell injury. Our findings revealed that salidroside attenuates MPP+ -induced neurotoxicity as evidenced by the increase in cell viability, and the decreases in the caspase-3 activity and apoptosis ratio. Simultaneously, salidroside pretreatment remarkably increased SIRT1 activity, SIRT1 mRNA and protein levels in MPP+ -treated SH-SY5Y cell. However, sirtinol, a SIRT1 activation inhibitor, significantly blocked the inhibitory effects of salidroside on MPP+ -induced cytotoxicity and apoptosis. In addition, salidroside abolished MPP+ -induced the production of reactive oxygen species (ROS), the up-regulation of NADPH oxidase 2 (NOX2) expression, the down-regulations of superoxide dismutase (SOD) activity and glutathione (GSH) level in SH-SY5Y cells, while these effects were also blocked by sirtinol. Finally, we found that the inhibition of salidroside on MPP+ -induced phosphorylation of p38, extracellular signal-regulated kinase (ERK) and c-Jun NH2-terminal kinase (JNK) were also reversed by sirtinol in SH-SY5Y cells. Taken together, these results indicated that SIRT1 contributes to the neuroprotection of salidroside against MPP+ -induced apoptosis and oxidative stress, in part through suppressing of mitogen-activated protein kinase (MAPK) pathways.
Collapse
Affiliation(s)
- Chun-Yang Wang
- Department of Science and Technology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhao-Nan Sun
- Department of General surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ming-Xin Wang
- Department of Otolaryngological, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chao Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
143
|
Razi S, Cogger VC, Kennerson M, Benson VL, McMahon AC, Blyth FM, Handelsman DJ, Seibel MJ, Hirani V, Naganathan V, Waite L, de Cabo R, Cumming RG, Le Couteur DG. SIRT1 Polymorphisms and Serum-Induced SIRT1 Protein Expression in Aging and Frailty: The CHAMP Study. J Gerontol A Biol Sci Med Sci 2017; 72:870-876. [PMID: 28329314 DOI: 10.1093/gerona/glx018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/21/2017] [Indexed: 01/02/2023] Open
Abstract
The nutrient sensing protein, SIRT1 influences aging and nutritional interventions such as caloric restriction in animals, however, the role of SIRT1 in human aging remains unclear. Here, the role of SIRT1 single-nucleotide polymorphisms (SNPs) and serum-induced SIRT1 protein expression (a novel assay that detects circulating factors that influence SIRT1 expression in vitro) were studied in the Concord Health and Ageing in Men Project (CHAMP), a prospective cohort of community dwelling men aged 70 years and older. Serum-induced SIRT1 expression was not associated with age or mortality, however participants within the lowest quintile were less likely to be frail (odds ratio (OR) 0.34, 95% confidence interval (CI) 0.17-0.69, N = 1,309). Serum-induced SIRT1 expression was associated with some markers of body composition and nutrition (height, weight, body fat and lean % mass, albumin, and cholesterol) but not disease. SIRT1 SNPs rs2273773, rs3740051, and rs3758391 showed no association with age, frailty, or mortality but were associated with weight, height, body fat and lean, and albumin levels. There were some weak associations between SIRT1 SNPs and arthritis, heart attack, deafness, and cognitive impairment. There was no association between SIRT1 SNPs and the serum-induced SIRT1 assay. SIRT1 SNPs and serum-induced SIRT1 expression in older men may be more closely associated with nutrition and body composition than aging and age-related conditions.
Collapse
Affiliation(s)
- Shajjia Razi
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia.,ANZAC Research Institute and
| | - Victoria C Cogger
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia.,ANZAC Research Institute and.,Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | | | - Vicky L Benson
- Department of Physiology, University of Auckland, New Zealand
| | - Aisling C McMahon
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia.,Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - Fiona M Blyth
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia
| | | | | | - Vasant Hirani
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia.,Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - Vasikaran Naganathan
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia
| | - Louise Waite
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland
| | - Robert G Cumming
- School of Public Health, University of Sydney, New South Wales, Australia
| | - David G Le Couteur
- Centre for Education and Research on Ageing and the Ageing and Alzheimers Institute, University of Sydney and Concord Hospital, New South Wales, Australia.,ANZAC Research Institute and.,Charles Perkins Centre, University of Sydney, New South Wales, Australia
| |
Collapse
|
144
|
Ma S, Feng J, Zhang R, Chen J, Han D, Li X, Yang B, Li X, Fan M, Li C, Tian Z, Wang Y, Cao F. SIRT1 Activation by Resveratrol Alleviates Cardiac Dysfunction via Mitochondrial Regulation in Diabetic Cardiomyopathy Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4602715. [PMID: 28883902 PMCID: PMC5572590 DOI: 10.1155/2017/4602715] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/31/2017] [Accepted: 06/06/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a major threat for diabetic patients. Silent information regulator 1 (SIRT1) has a regulatory effect on mitochondrial dynamics, which is associated with DCM pathological changes. Our study aims to investigate whether resveratrol, a SRIT1 activator, could exert a protective effect against DCM. METHODS AND RESULTS Cardiac-specific SIRT1 knockout (SIRT1KO) mice were generated using Cre-loxP system. SIRT1KO mice displayed symptoms of DCM, including cardiac hypertrophy and dysfunction, insulin resistance, and abnormal glucose metabolism. DCM and SIRT1KO hearts showed impaired mitochondrial biogenesis and function, while SIRT1 activation by resveratrol reversed this in DCM mice. High glucose caused increased apoptosis, impaired mitochondrial biogenesis, and function in cardiomyocytes, which was alleviated by resveratrol. SIRT1 deletion by both SIRT1KO and shRNA abolished the beneficial effects of resveratrol. Furthermore, the function of SIRT1 is mediated via the deacetylation effect on peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), thus inducing increased expression of nuclear respiratory factor 1 (NRF-1), NRF-2, estrogen-related receptor-α (ERR-α), and mitochondrial transcription factor A (TFAM). CONCLUSIONS Cardiac deletion of SIRT1 caused phenotypes resembling DCM. Activation of SIRT1 by resveratrol ameliorated cardiac injuries in DCM through PGC-1α-mediated mitochondrial regulation. Collectively, SIRT1 may serve as a potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Sai Ma
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jing Feng
- Department of Emergency Medicine, Jinling Hospital, Nanjing, Jiangsu, China
| | - Ran Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jiangwei Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dong Han
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bo Yang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Xiujuan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Miaomiao Fan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zuhong Tian
- Department of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yabin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
145
|
Lemos V, de Oliveira RM, Naia L, Szegö É, Ramos E, Pinho S, Magro F, Cavadas C, Rego AC, Costa V, Outeiro TF, Gomes P. The NAD+-dependent deacetylase SIRT2 attenuates oxidative stress and mitochondrial dysfunction and improves insulin sensitivity in hepatocytes. Hum Mol Genet 2017; 26:4105-4117. [DOI: 10.1093/hmg/ddx298] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/23/2017] [Indexed: 01/11/2023] Open
|
146
|
The Role of Ghrelin and Ghrelin Signaling in Aging. Int J Mol Sci 2017; 18:ijms18071511. [PMID: 28704966 PMCID: PMC5536001 DOI: 10.3390/ijms18071511] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/04/2017] [Accepted: 07/07/2017] [Indexed: 01/20/2023] Open
Abstract
With our aging society, more people hope for a long and healthy life. In recent years, researchers have focused on healthy longevity factors. In particular, calorie restriction delays aging, reduces mortality, and extends life. Ghrelin, which is secreted during fasting, is well known as an orexigenic peptide. Because ghrelin is increased by caloric restriction, ghrelin may play an important role in the mechanism of longevity mediated by calorie restriction. In this review, we will discuss the role of orexigenic peptides with a particular focus on ghrelin. We conclude that the ghrelin-growth hormone secretagogue-R signaling pathway may play an important role in the anti-aging mechanism.
Collapse
|
147
|
Zhou L, Wang SI, Moon YJ, Kim KM, Lee KB, Park BH, Jang KY, Kim JR. Overexpression of SIRT1 prevents hypoxia-induced apoptosis in osteoblast cells. Mol Med Rep 2017; 16:2969-2975. [DOI: 10.3892/mmr.2017.6917] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 04/28/2017] [Indexed: 11/06/2022] Open
|
148
|
Li Y, Meng X, Wang W, Liu F, Hao Z, Yang Y, Zhao J, Yin W, Xu L, Zhao R, Hu J. Cardioprotective Effects of SIRT6 in a Mouse Model of Transverse Aortic Constriction-Induced Heart Failure. Front Physiol 2017; 8:394. [PMID: 28659816 PMCID: PMC5468374 DOI: 10.3389/fphys.2017.00394] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/26/2017] [Indexed: 01/23/2023] Open
Abstract
SIRT6, a member of the NAD (+)-dependent class III deacetylase sirtuin family, plays important roles in the maintenance of cardiovascular homeostasis. Telomere shortening is a risk factor for age-associated diseases, including heart disease. In the present study, we investigated the role of SIRT6 and telomerase in a mouse model of transverse aortic constriction (TAC)-induced heart failure. SIRT6, telomerase reverse transcriptase (TERT), and telomere repeat binding factor (TRF)-1 were significantly downregulated in TAC mice compared with their expression in sham-operated mice. Lentiviral vector-mediated overexpression of SIRT6 upregulated TERT and TRF1 and increased the survival of mice after TAC. Echocardiography and hemodynamic measurements as well as histological analyses indicated that SIRT6 overexpression attenuated TAC-induced heart dysfunction and decreased TAC-induced cardiac inflammatory responses, reducing cardiac fibrosis and decreasing infarct size. Taken together, our findings indicate that SIRT6 protects the myocardium against damage and this effect may be mediated by the modulation of telomeres. Our findings linking SIRT6 and telomere integrity in the heart warrant further investigation into the underlying mechanisms and support SIRT6 as a promising therapeutic target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yongming Li
- Department of Cardiology, Baotou Central HospitalBaotou, China
| | - Xianda Meng
- Department of Cardiology, Dalian (Municipal) Friendship HospitalDalian, China
| | - Wenguang Wang
- Department of Cardiology, Baotou Central HospitalBaotou, China
| | - Fu Liu
- Department of Cardiology, Baotou Central HospitalBaotou, China
| | - Zhiru Hao
- Department of Cardiology, Baotou Central HospitalBaotou, China
| | - Yang Yang
- Department of Cardiology, Baotou Central HospitalBaotou, China
| | - Jinbo Zhao
- Department of Cardiology, Baotou Central HospitalBaotou, China
| | - Wensi Yin
- Department of Institution of Interventional and Vascular Surgery, Tongji UniversityShanghai, China
| | - Lijuan Xu
- Department of Institution of Interventional and Vascular Surgery, Tongji UniversityShanghai, China
| | - Ruiping Zhao
- Department of Cardiology, Baotou Central HospitalBaotou, China.,Tanslational Medicine Center, Baotou Central HospitalBaotou, China
| | - Jiang Hu
- Tanslational Medicine Center, Baotou Central HospitalBaotou, China
| |
Collapse
|
149
|
Al-Waili N, Al-Waili H, Al-Waili T, Salom K. Natural antioxidants in the treatment and prevention of diabetic nephropathy; a potential approach that warrants clinical trials. Redox Rep 2017; 22:99-118. [PMID: 28276289 PMCID: PMC6837693 DOI: 10.1080/13510002.2017.1297885] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy is the major cause of end-stage renal disease and effective and new therapeutic approaches are needed in diabetic nephropathy and chronic kidney diseases. Oxidative stress and inflammatory process are important factors contributing to kidney damage by increasing production of oxidants. KEAP1/Nrf2/ARE pathway regulates the transcription of many antioxidant genes and modulation of the pathway up regulates antioxidants. NFB controls the expression of genes involved in the inflammatory response. Natural substances have antioxidant and anti-inflammatory activities and have an impact on NFB and KEAP1/Nrf2/ARE pathways. The preclinical studies explored the effectiveness of whole herbs, plants or seeds and their active ingredients in established diabetic nephropathy. They ameliorate oxidative stress induced kidney damage, enhance antioxidant system, and decrease inflammatory process and fibrosis; most likely by activating KEAP1/Nrf2/ARE pathway and by deactivating NFB pathway. Whole natural products contain balanced antioxidants that might work synergistically to induce beneficial therapeutic outcome. In this context, more clinical studies involving whole plants or herbal products or mixtures of different herbs and plants and their active ingredients might change our strategies for the management of diabetic nephropathy. The natural products might be useful as preventive interventions and studies are required in this field.
Collapse
Affiliation(s)
- Noori Al-Waili
- New York Medical Care for Nephrology, Al-Waili Foundation for Science, New York, USA
| | - Hamza Al-Waili
- New York Medical Care for Nephrology, Al-Waili Foundation for Science, New York, USA
| | - Thia Al-Waili
- New York Medical Care for Nephrology, Al-Waili Foundation for Science, New York, USA
| | - Khelod Salom
- New York Medical Care for Nephrology, Al-Waili Foundation for Science, New York, USA
| |
Collapse
|
150
|
Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG, de Cabo R. Pharmacological Strategies to Retard Cardiovascular Aging. Circ Res 2017; 118:1626-42. [PMID: 27174954 DOI: 10.1161/circresaha.116.307475] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/08/2016] [Indexed: 01/10/2023]
Abstract
Aging is the major risk factor for cardiovascular diseases, which are the leading cause of death in the United States. Traditionally, the effort to prevent cardiovascular disease has been focused on addressing the conventional risk factors, including hypertension, hyperglycemia, hypercholesterolemia, and high circulating levels of triglycerides. However, recent preclinical studies have identified new approaches to combat cardiovascular disease. Calorie restriction has been reproducibly shown to prolong lifespan in various experimental model animals. This has led to the development of calorie restriction mimetics and other pharmacological interventions capable to delay age-related diseases. In this review, we will address the mechanistic effects of aging per se on the cardiovascular system and focus on the prolongevity benefits of various therapeutic strategies that support cardiovascular health.
Collapse
Affiliation(s)
- Irene Alfaras
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Clara Di Germanio
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Michel Bernier
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Anna Csiszar
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Zoltan Ungvari
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Edward G Lakatta
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Rafael de Cabo
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.).
| |
Collapse
|