201
|
Abstract
SIGNIFICANCE Developing evidence in the literature suggests that sirtuin 5 (SIRT5) may be involved in metabolic reprogramming, an emerging hallmark of cancer by which neoplastic cells reconfigure their metabolism to support the anabolic demands of rapid cell division. SIRT5 is one of the seven members of the nicotinamide adenine dinucleotide-dependent sirtuin family of lysine deacetylases. It removes succinyl, malonyl, and glutaryl groups from protein targets within the mitochondrial matrix and other subcellular compartments. SIRT5 substrates include a number of proteins integral to metabolism. Recent Advances: New work has begun to elucidate the roles of SIRT5 in glycolysis, tricarboxylic acid cycle, fatty acid oxidation, nitrogen metabolism, pentose phosphate pathway, antioxidant defense, and apoptosis. CRITICAL ISSUES In this study, we summarize biological functions of SIRT5 reported in normal tissues and in cancer and discuss potential mechanisms whereby SIRT5 may impact tumorigenesis, particularly focusing on its reported roles in metabolic reprogramming. Finally, we review current efforts to target SIRT5 pharmacologically. FUTURE DIRECTIONS The biological significance of SIRT5 has been elucidated in the context of only an extremely small fraction of its targets and interactors. There is no doubt that further studies in this area will provide a wealth of insights into functions of SIRT5 and its targets in normal and neoplastic cells. Antioxid. Redox Signal. 28, 677-690.
Collapse
Affiliation(s)
| | - Angela H. Guo
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - David B. Lombard
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
202
|
Abstract
SIGNIFICANCE Extranuclear sirtuins in cytosol (SIRT2) and mitochondria (SIRT3, SIRT4, and SIRT5) are key regulators of metabolic enzymes and the antioxidative defense mechanisms. They play an important role in the adjustment of metabolic pathways in alterations of the nutritional status. Recent Advances: Recent studies have shown that in addition to lysine deacetylation, sirtuins catalyze several different lysine deacylation reactions, removal of lipid modifications, and adenosine diphosphate-ribosylation. Large-scale studies have revealed hundreds of target proteins regulated by different sirtuin modifications. CRITICAL ISSUES Sensing of the metabolic state and regulation of the sirtuin function and expression are critical components of the machinery, optimizing cellular functions in the switch from fed to fasting condition. Overfeeding, obesity, and metabolic diseases cause metabolic stress that dysregulates the sirtuins, which may play a role in the pathogenesis and complications of metabolic diseases such as type 2 diabetes, fatty liver disease, and cardiac diseases. In the current review, we will discuss the significance of the extranuclear sirtuins as metabolic regulators and in protection against the reactive oxygen species, and also how these sirtuins are regulated by metabolic status and their putative role in metabolic diseases. FUTURE DIRECTIONS To efficiently utilize sirtuins as drug targets for treatment of the metabolic diseases, better understanding of the sirtuin functions, targets, regulation, and cross talk is needed. Furthermore, more studies in humans are needed to confirm the many observations mainly made in animal and cell models so far. Antioxid. Redox Signal. 28, 662-676.
Collapse
Affiliation(s)
- Mahmoud-Sobhy Elkhwanky
- 1 Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu , Oulu, Finland .,2 Medical Research Center Oulu, Oulu University Hospital and University of Oulu , Oulu, Finland
| | - Jukka Hakkola
- 1 Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu , Oulu, Finland .,2 Medical Research Center Oulu, Oulu University Hospital and University of Oulu , Oulu, Finland
| |
Collapse
|
203
|
Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid Redox Signal 2018; 28:643-661. [PMID: 28891317 PMCID: PMC5824489 DOI: 10.1089/ars.2017.7290] [Citation(s) in RCA: 544] [Impact Index Per Article: 77.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Antioxidant and redox signaling (ARS) events are regulated by critical molecules that modulate antioxidants, reactive oxygen species (ROS) or reactive nitrogen species (RNS), and/or oxidative stress within the cell. Imbalances in these molecules can disturb cellular functions to become pathogenic. Sirtuins serve as important regulators of ARS in cells. Recent Advances: Sirtuins (SIRTs 1-7) are a family of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases with the ability to deacetylate histone and nonhistone targets. Recent studies show that sirtuins modulate the regulation of a variety of cellular processes associated with ARS. SIRT1, SIRT3, and SIRT5 protect the cell from ROS, and SIRT2, SIRT6, and SIRT7 modulate key oxidative stress genes and mechanisms. Interestingly, SIRT4 has been shown to induce ROS production and has antioxidative roles as well. CRITICAL ISSUES A complete understanding of the roles of sirtuins in redox homeostasis of the cell is very important to understand the normal functioning as well as pathological manifestations. In this review, we have provided a critical discussion on the role of sirtuins in the regulation of ARS. We have also discussed mechanistic interactions among different sirtuins. Indeed, a complete understanding of sirtuin biology could be critical at multiple fronts. FUTURE DIRECTIONS Sirtuins are emerging to be important in normal mammalian physiology and in a variety of oxidative stress-mediated pathological situations. Studies are needed to dissect the mechanisms of sirtuins in maintaining redox homeostasis. Efforts are also required to assess the targetability of sirtuins in the management of redox-regulated diseases. Antioxid. Redox Signal. 28, 643-661.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | | | - Nicholas J Mack
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| |
Collapse
|
204
|
SIRT2-mediated FOXO3a deacetylation drives its nuclear translocation triggering FasL-induced cell apoptosis during renal ischemia reperfusion. Apoptosis 2018; 22:519-530. [PMID: 28078537 DOI: 10.1007/s10495-016-1341-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We have found that Fas/FasL-mediated "extrinsic" pathway promoted cell apoptosis induced by renal ischemic injury. This study is to elucidate the upstream mechanism regulating FasL-induced extrinsic pathway during renal ischemia/reperfusion. Results demonstrated that when SIRT2 was activated by renal ischemia/reperfusion, activated SIRT2 could bind to and deacetylate FOXO3a, promoting FOXO3a nuclear translocation which resulted in an increase of nuclear FOXO3a along with FasL expression and activation of caspase8 and caspase3, triggering cell apoptosis during renal ischemia/reperfusion. The administration of SIRT2 inhibitor AGK2 prior to renal ischemia decreased significantly the number of apoptotic renal tubular cells and alleviated ultrastructure injury. These results indicate that inhibition of FOXO3a deacetylation might be a promising therapeutic approach for renal ischemia /reperfusion injury.
Collapse
|
205
|
Liu T, Yang W, Pang S, Yu S, Yan B. Functional genetic variants within the SIRT2 gene promoter in type 2 diabetes mellitus. Diabetes Res Clin Pract 2018; 137:200-207. [PMID: 29371109 DOI: 10.1016/j.diabres.2018.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
AIMS Type 2 diabetes mellitus (T2D) is a common and complex metabolic diseases caused by interactions between environmental and genetic factors. Genome-wide association studies have identified more than 80 common genetic variants for T2D, which account for only ∼10% of the heritability of T2D cases. SIRT2, a member of NAD(+)-dependent class III deacetylases, is involved in genomic stability, metabolism, inflammation, oxidative stress and autophagy. In maintaining metabolic homeostasis, SIRT2 regulates adipocyte differentiation, fatty acid oxidation, gluconeogenesis, and insulin sensitivity. Thus, we hypothesized that DNA sequence variants (DSVs) in SIRT2 gene promoter may change SIRT2 levels, contributing to T2D. METHODS SIRT2 gene promoter was genetically and functionally analyzed in large cohorts of T2D patients (n = 365) and ethnic-matched controls (n = 358). RESULTS A total of 18 DSVs, including 5 SNPs, were identified in this study. Four novel heterozygous DSVs (g.38900912G > T, g.38900561C > T, g.38900359C > T and g.38900237G > A) were identified in four T2D patients, three of which (g.38900912G > T, g.38900359C > T and g.38900237G > A) significantly increased the transcriptional activity of the SIRT2 gene promoter in cultured pancreatic beta cells (P < .01). Seven novel heterozygous DSVs were only found in controls, and one heterozygous deletion DSV and five SNPs were found in both T2D patients and controls, which did not significantly affect SIRT2 gene promoter activity (P > .05). CONCLUSIONS Our findings suggested that the DSVs may increase SIRT2 gene promoter activity and SIRT2 levels, contributing to T2D development as a risk factor.
Collapse
Affiliation(s)
- Tingting Liu
- College of Clinical Medicine, Jining Medical University, Jining, Shandong 272100, China
| | - Wentao Yang
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Shipeng Yu
- Division of Endocrinology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
206
|
Keskin-Aktan A, Akbulut KG, Yazici-Mutlu Ç, Sonugur G, Ocal M, Akbulut H. The effects of melatonin and curcumin on the expression of SIRT2, Bcl-2 and Bax in the hippocampus of adult rats. Brain Res Bull 2018; 137:306-310. [DOI: 10.1016/j.brainresbull.2018.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/10/2017] [Accepted: 01/05/2018] [Indexed: 01/15/2023]
|
207
|
SIRT2 regulates oxidative stress-induced cell death through deacetylation of c-Jun NH 2-terminal kinase. Cell Death Differ 2018; 25:1638-1656. [PMID: 29449643 DOI: 10.1038/s41418-018-0069-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/06/2017] [Accepted: 01/15/2018] [Indexed: 01/08/2023] Open
Abstract
c-Jun NH2-terminal kinases (JNKs) are responsive to stress stimuli and their activation regulate key cellular functions, including cell survival, growth, differentiation and aging. Previous studies demonstrate that activation of JNK requires dual phosphorylation by the mitogen-activated protein kinase kinases. However, other post-translational mechanisms involved in regulating the activity of JNK have been poorly understood. In this work, we studied the functional significance of reversible lysine acetylation in regulating the kinase activity of JNK. We found that the acetyl transferase p300 binds to, acetylates and inhibits kinase activity of JNK. Using tandem mass spectrometry, molecular modelling and molecular dynamics simulations, we found that acetylation of JNK at Lys153 would hinder the stable interactions of the negatively charged phosphates and prevent the adenosine binding to JNK. Our screening for the deacetylases found SIRT2 as a deacetylase for JNK. Mechanistically, SIRT2-dependent deacetylation enhances ATP binding and enzymatic activity of JNK towards c-Jun. Furthermore, SIRT2-mediated deacetylation favours the phosphorylation of JNK by MKK4, an upstream kinase. Our results indicate that deacetylation of JNK by SIRT2 promotes oxidative stress-induced cell death. Conversely, SIRT2 inhibition attenuates H2O2-mediated cell death in HeLa cells. SIRT2-deficient (SIRT2-KO) mice exhibit increased acetylation of JNK, which is associated with markedly reduced catalytic activity of JNK in the liver. Interestingly, SIRT2-KO mice were resistant to acetaminophen-induced liver toxicity. SIRT2-KO mice show lower cell death, minimal degenerative changes, improved liver function and survival following acetaminophen treatment. Overall, our work identifies SIRT2-mediated deacetylation of JNK as a critical regulator of cell survival during oxidative stress.
Collapse
|
208
|
Tatone C, Di Emidio G, Barbonetti A, Carta G, Luciano AM, Falone S, Amicarelli F. Sirtuins in gamete biology and reproductive physiology: emerging roles and therapeutic potential in female and male infertility. Hum Reprod Update 2018; 24:267-289. [DOI: 10.1093/humupd/dmy003] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/19/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Carla Tatone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Gynecology Unit, Reproductive Service, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy
| | - Giovanna Di Emidio
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Gynecology Unit, Reproductive Service, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy
| | | | - Gaspare Carta
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Gynecology Unit, Reproductive Service, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy
| | - Alberto M Luciano
- Department of Health, Animal Science and Food Safety, Reproductive and Developmental Biology Laboratory, University of Milan, 20133 Milan, Italy
| | - Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Institute of Translational Pharmacology (IFT), CNR, 67100 L’Aquila, Italy
| |
Collapse
|
209
|
Yadav RK, Chauhan AS, Zhuang L, Gan B. FoxO transcription factors in cancer metabolism. Semin Cancer Biol 2018; 50:65-76. [PMID: 29309929 DOI: 10.1016/j.semcancer.2018.01.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/28/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022]
Abstract
FoxO transcription factors serve as the central regulator of cellular homeostasis and are tumor suppressors in human cancers. Recent studies have revealed that, besides their classic functions in promoting cell death and inducing cell cycle arrest, FoxOs also regulate cancer metabolism, an emerging hallmark of cancer. In this review, we summarize the regulatory mechanisms employed to control FoxO activities in the context of cancer biology, and discuss FoxO function in metabolism reprogramming in cancer and interaction with other key cancer metabolism pathways. A deeper understanding of FoxOs in cancer metabolism may reveal novel therapeutic opportunities in cancer treatment.
Collapse
Affiliation(s)
- Raj Kumar Yadav
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Anoop Singh Chauhan
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | - Li Zhuang
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; Department of Molecular and Cellular Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| |
Collapse
|
210
|
Hornedo-Ortega R, Da Costa G, Cerezo AB, Troncoso AM, Richard T, Garcia-Parrilla MC. In Vitro Effects of Serotonin, Melatonin, and Other Related Indole Compounds on Amyloid-β Kinetics and Neuroprotection. Mol Nutr Food Res 2018; 62. [PMID: 29131485 DOI: 10.1002/mnfr.201700383] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/25/2017] [Indexed: 11/08/2022]
Abstract
SCOPE Amyloid-β peptide is the main component of senile plaques in Alzheimer's disease. The inhibition of amyloid-β peptide assembly, the destabilization of amyloid-β peptide aggregates, and the decrease of its cytotoxicity for the prevention of neuronal death are considered neuroprotective effects. In this work, the protective effects against amyloid-β peptide aggregation and cytotoxicity of eight indolic compounds are evaluated: tryptophan, tryptamine, serotonin, tryptophol, N-acetylserotonin, 3-indoleacetic acid, tryptophan ethyl ester, and melatonin. METHODS AND RESULTS Thioflavin T spectroscopic assay, transmission electron microscopy, western blotting, circular dichroism, NMR, cell viability (thiazolyl blue tetrazolium bromide assay), quantitative PCR, and heme oxygenase activity are used. Serotonin is the most effective compound for inhibiting amyloid-β peptide aggregation. Almost all the indolic compounds tested prevent amyloid-β peptide-induced and increase cell viability, being between 9 and 25%. Melatonin and serotonin are the most active. Moreover, serotonin increased the expression of SIRT-1 and 2, heat shock protein 70, and heme oxygenase activity, this being a possible mechanism underlying the observed neuroprotective effect. CONCLUSION Melatonin and other related indolic compounds, mainly serotonin, show an inhibitory and destabilizing effect on amyloid-β peptide fibril formation and they possess neuroprotective properties related to the vitagenes system.
Collapse
Affiliation(s)
- Ruth Hornedo-Ortega
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Grégory Da Costa
- ISVV, Unité de Recherche Oenologie, Université de Bordeaux, Villenave d'Ornon, France
| | - Ana B Cerezo
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Ana M Troncoso
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Tristan Richard
- ISVV, Unité de Recherche Oenologie, Université de Bordeaux, Villenave d'Ornon, France
| | - M Carmen Garcia-Parrilla
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
211
|
Song J, Yang B, Jia X, Li M, Tan W, Ma S, Shi X, Feng L. Distinctive Roles of Sirtuins on Diabetes, Protective or Detrimental? Front Endocrinol (Lausanne) 2018; 9:724. [PMID: 30559718 PMCID: PMC6284472 DOI: 10.3389/fendo.2018.00724] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022] Open
Abstract
Dysregulation of metabolic pathways leads to type 2 diabetes, characteristic of high glucose concentration caused by insulin resistance. The histone deacetylases sirtuins exhibit remarkable enzymatic activities. Accumulating evidence indicates that sirtuins can be pharmacologically activated to ameliorate diabetes. Here, we evaluated different roles of sirtuins (SIRT1-SIRT7) in diabetes progression and described their involvement in metabolic pathways of skeletal muscle, adipose tissue and liver. The nuclear sirtuins, SIRT1, SIRT6, and SIRT7, regulate the activity of key transcription factors and cofactors in almost all tissues with the cellular responses to energy demands. The mitochondrial sirtuins, SIRT3, SIRT4, and SIRT5, regulate the activity of mitochondrial enzymes in response to fasting and calorie restriction. Moreover, genetic polymorphisms of SIRT1 and SIRT2 have been reported to associate with diabetes development. It's worth noting that SIRT1, SIRT2, SIRT3, and SIRT6 are positive regulators of insulin resistance in most cases. In the opposite, SIRT4 and SIRT7 inhibit insulin secretion and fatty acid oxidation. Identification of SIRT1 activators for diabetes has gained wide attention, such as metformin, resveratrol, and resveratrol derivatives. Randomized, prospective, and large-scale clinical trials are warrant to uncover the responsibilities of SIRTs modulators on diabetes progress.
Collapse
Affiliation(s)
- Jie Song
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mingyu Li
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Tan
- Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shitang Ma
- Life and Health college, Anhui Science and Technology University, Fengyang, China
| | - Xinhong Shi
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- *Correspondence: Liang Feng
| |
Collapse
|
212
|
Abstract
The mammalian Sirtuins (SIRT1-7) are an evolutionarily conserved family of NAD+-dependent deacylase and mono-ADP-ribosyltransferase. Sirtuins display distinct subcellular localizations and functions and are involved in cell survival, senescence, metabolism and genome stability. Among the mammalian Sirtuins, SIRT1 and SIRT6 have been thoroughly investigated and have prominent metabolic regulatory roles. Moreover, SIRT1 and SIRT6 have been implicated in obesity, insulin resistance, type 2 diabetes mellitus (T2DM), fatty liver disease and cardiovascular diseases. However, the roles of other Sirtuins are not fully understood. Recent studies have shown that these Sirtuins also play important roles in inflammation, mitochondrial dysfunction, and energy metabolism. Insulin resistance is the critical pathological trait of obesity and metabolic syndrome as well as the core defect in T2DM. Accumulating clinical and experimental animal evidence suggests the potential roles of the remaining Sirtuins in the regulation of insulin resistance through diverse biological mechanisms. In this review, we summarize recent advances in the understanding of the functions of Sirtuins in various insulin resistance-associated physiological processes, including inflammation, mitochondrial dysfunction, the insulin signaling pathway, glucose, and lipid metabolism. In addition, we highlight the important gaps that must be addressed in this field.
Collapse
Affiliation(s)
- Shuang Zhou
- Internal Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaoqiang Tang
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Hou-Zao Chen ;
| |
Collapse
|
213
|
Pomatto LCD, Davies KJA. The role of declining adaptive homeostasis in ageing. J Physiol 2017; 595:7275-7309. [PMID: 29028112 PMCID: PMC5730851 DOI: 10.1113/jp275072] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
Adaptive homeostasis is "the transient expansion or contraction of the homeostatic range for any given physiological parameter in response to exposure to sub-toxic, non-damaging, signalling molecules or events, or the removal or cessation of such molecules or events" (Davies, 2016). Adaptive homeostasis enables biological systems to make continuous short-term adjustments for optimal functioning despite ever-changing internal and external environments. Initiation of adaptation in response to an appropriate signal allows organisms to successfully cope with much greater, normally toxic, stresses. These short-term responses are initiated following effective signals, including hypoxia, cold shock, heat shock, oxidative stress, exercise-induced adaptation, caloric restriction, osmotic stress, mechanical stress, immune response, and even emotional stress. There is now substantial literature detailing a decline in adaptive homeostasis that, unfortunately, appears to manifest with ageing, especially in the last third of the lifespan. In this review, we present the hypothesis that one hallmark of the ageing process is a significant decline in adaptive homeostasis capacity. We discuss the mechanistic importance of diminished capacity for short-term (reversible) adaptive responses (both biochemical and signal transduction/gene expression-based) to changing internal and external conditions, for short-term survival and for lifespan and healthspan. Studies of cultured mammalian cells, worms, flies, rodents, simians, apes, and even humans, all indicate declining adaptive homeostasis as a potential contributor to age-dependent senescence, increased risk of disease, and even mortality. Emerging work points to Nrf2-Keap1 signal transduction pathway inhibitors, including Bach1 and c-Myc, both of whose tissue concentrations increase with age, as possible major causes for age-dependent loss of adaptive homeostasis.
Collapse
Affiliation(s)
- Laura C. D. Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
| | - Kelvin J. A. Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
- Molecular and Computational Biology Program, Department of Biological Sciences of the Dornsife College of LettersArts & Sciences: the University of Southern CaliforniaLos AngelesCA 90089‐0191USA
| |
Collapse
|
214
|
Drake LE, Springer MZ, Poole LP, Kim CJ, Macleod KF. Expanding perspectives on the significance of mitophagy in cancer. Semin Cancer Biol 2017; 47:110-124. [PMID: 28450176 PMCID: PMC5654704 DOI: 10.1016/j.semcancer.2017.04.008] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023]
Abstract
Mitophagy is a selective mode of autophagy in which mitochondria are specifically targeted for degradation at the autophagolysosome. Mitophagy is activated by stresses such as hypoxia, nutrient deprivation, DNA damage, inflammation and mitochondrial membrane depolarization and plays a role in maintaining mitochondrial integrity and function. Defects in mitophagy lead to mitochondrial dysfunction that can affect metabolic reprogramming in response to stress, alter cell fate determination and differentiation, which in turn affects disease incidence and etiology, including cancer. Here, we discuss how different mitophagy adaptors and modulators, including Parkin, BNIP3, BNIP3L, p62/SQSTM1 and OPTN, are regulated in response to physiological stresses and deregulated in cancers. Additionally, we explore how these different mitophagy control pathways coordinate with each other. Finally, we review new developments in understanding how mitophagy affects stemness, cell fate determination, inflammation and DNA damage responses that are relevant to understanding the role of mitophagy in cancer.
Collapse
Affiliation(s)
- Lauren E Drake
- The Ben May Department for Cancer Research, The University of Chicago, USA
| | - Maya Z Springer
- The Ben May Department for Cancer Research, The University of Chicago, USA; The Committee on Cancer Biology, The University of Chicago, USA
| | - Logan P Poole
- The Ben May Department for Cancer Research, The University of Chicago, USA; The Committee on Cancer Biology, The University of Chicago, USA
| | - Casey J Kim
- The Ben May Department for Cancer Research, The University of Chicago, USA
| | - Kay F Macleod
- The Ben May Department for Cancer Research, The University of Chicago, USA; The Committee on Cancer Biology, The University of Chicago, USA.
| |
Collapse
|
215
|
Fourcade S, Morató L, Parameswaran J, Ruiz M, Ruiz‐Cortés T, Jové M, Naudí A, Martínez‐Redondo P, Dierssen M, Ferrer I, Villarroya F, Pamplona R, Vaquero A, Portero‐Otín M, Pujol A. Loss of SIRT2 leads to axonal degeneration and locomotor disability associated with redox and energy imbalance. Aging Cell 2017; 16:1404-1413. [PMID: 28984064 PMCID: PMC5676070 DOI: 10.1111/acel.12682] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2017] [Indexed: 12/13/2022] Open
Abstract
Sirtuin 2 (SIRT2) is a member of a family of NAD+‐dependent histone deacetylases (HDAC) that play diverse roles in cellular metabolism and especially for aging process. SIRT2 is located in the nucleus, cytoplasm, and mitochondria, is highly expressed in the central nervous system (CNS), and has been reported to regulate a variety of processes including oxidative stress, genome integrity, and myelination. However, little is known about the role of SIRT2 in the nervous system specifically during aging. Here, we show that middle‐aged, 13‐month‐old mice lacking SIRT2 exhibit locomotor dysfunction due to axonal degeneration, which was not present in young SIRT2 mice. In addition, these Sirt2−/− mice exhibit mitochondrial depletion resulting in energy failure, and redox dyshomeostasis. Our results provide a novel link between SIRT2 and physiological aging impacting the axonal compartment of the central nervous system, while supporting a major role for SIRT2 in orchestrating its metabolic regulation. This underscores the value of SIRT2 as a therapeutic target in the most prevalent neurodegenerative diseases that undergo with axonal degeneration associated with redox and energetic dyshomeostasis.
Collapse
Affiliation(s)
- Stéphane Fourcade
- Neurometabolic Diseases Laboratory Institute of Neuropathology IDIBELL Barcelona Spain
- CIBERER U759 Center for Biomedical Research on Rare Diseases Barcelona Spain
| | - Laia Morató
- Neurometabolic Diseases Laboratory Institute of Neuropathology IDIBELL Barcelona Spain
- CIBERER U759 Center for Biomedical Research on Rare Diseases Barcelona Spain
| | - Janani Parameswaran
- Neurometabolic Diseases Laboratory Institute of Neuropathology IDIBELL Barcelona Spain
- CIBERER U759 Center for Biomedical Research on Rare Diseases Barcelona Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory Institute of Neuropathology IDIBELL Barcelona Spain
- CIBERER U759 Center for Biomedical Research on Rare Diseases Barcelona Spain
| | - Tatiana Ruiz‐Cortés
- Biogenesis Research Group Agrarian Sciences Faculty University of Antioquia Medellin Colombia
| | - Mariona Jové
- Experimental Medicine Department University of Lleida‐IRBLleida Lleida Spain
| | - Alba Naudí
- Experimental Medicine Department University of Lleida‐IRBLleida Lleida Spain
| | - Paloma Martínez‐Redondo
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC) Bellvitge Biomedical Research Institute (IDIBELL) 08908 L'Hospitalet de Llobregat, Barcelona Spain
| | - Mara Dierssen
- Cellular & Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation The Barcelona Institute of Science and Technology Barcelona Spain
- Department of Experimental and Health Sciences Universidad Pompeu Fabra Barcelona Spain
- CIBERER U716 Center for Biomedical Research on Rare Diseases Barcelona Spain
| | - Isidre Ferrer
- Institute of Neuropathology University of Barcelona L'Hospitalet de Llobregat, Barcelona Spain
- Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED) ISCIII Madrid Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biology University of Barcelona Av. Diagonal 643 08028 Barcelona, Catalonia Spain
- The Institute of Biomedicine of the University of Barcelona (IBUB) Barcelona Spain
- Center for Biomedical Research on Physiopathology of Obesity and Nutrition (CIBEROBN) Barcelona Spain
| | - Reinald Pamplona
- Experimental Medicine Department University of Lleida‐IRBLleida Lleida Spain
| | - Alejandro Vaquero
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC) Bellvitge Biomedical Research Institute (IDIBELL) 08908 L'Hospitalet de Llobregat, Barcelona Spain
| | - Manel Portero‐Otín
- Experimental Medicine Department University of Lleida‐IRBLleida Lleida Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory Institute of Neuropathology IDIBELL Barcelona Spain
- CIBERER U759 Center for Biomedical Research on Rare Diseases Barcelona Spain
- Catalan Institution of Research and Advanced Studies (ICREA) Barcelona Spain
| |
Collapse
|
216
|
Hou T, Li Z, Zhao Y, Zhu WG. Mechanisms controlling the anti-neoplastic functions of FoxO proteins. Semin Cancer Biol 2017; 50:101-114. [PMID: 29155239 DOI: 10.1016/j.semcancer.2017.11.007] [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: 06/01/2017] [Revised: 10/18/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023]
Abstract
The Forkhead box O (FoxO) proteins comprise a family of evolutionarily conserved transcription factors that predominantly function as tumor suppressors. These proteins assume diverse roles in the cellular anti-neoplastic response, including regulation of apoptosis and autophagy, cancer metabolism, cell-cycle arrest, oxidative stress and the DNA damage response. More recently, FoxO proteins have been implicated in cancer immunity and cancer stem-cell (CSC) homeostasis. Interestingly, in some sporadic sub-populations, FoxO protein function may also be manipulated by factors such as β-catenin whereby they instead can facilitate cancer progression via maintenance of CSC properties or promoting drug resistance or metastasis and invasion. This review highlights the essential biological functions of FoxOs and explores the areas that may be exploited in FoxO protein signaling pathways in the development of novel cancer therapeutic agents.
Collapse
Affiliation(s)
- Tianyun Hou
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhiming Li
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ying Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei-Guo Zhu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
217
|
Zhao G, Wang H, Xu C, Wang P, Chen J, Wang P, Sun Z, Su Y, Wang Z, Han L, Tong T. SIRT6 delays cellular senescence by promoting p27Kip1 ubiquitin-proteasome degradation. Aging (Albany NY) 2017; 8:2308-2323. [PMID: 27794562 PMCID: PMC5115890 DOI: 10.18632/aging.101038] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/04/2016] [Indexed: 12/15/2022]
Abstract
Sirtuin6(SIRT6) has been implicated as a key factor in aging and aging-related diseases. However, the role of SIRT6 in cellular senescence has not been fully understood. Here, we show that SIRT6 repressed the expression of p27Kip1 (p27) in cellular senescence. The expression of SIRT6 was reduced during cellular senescence, whereas enforced SIRT6 expression promoted cell proliferation and antagonized cellular senescence. In addition, we demonstrated that SIRT6 promoted p27 degradation by proteasome and SIRT6 decreased the acetylation level and protein half-life of p27. p27 acetylation increased its protein stability. Furthermore, SIRT6 directly interacted with p27. Importantly, p27 was strongly acetylated and had a prolonged protein half-life with the reduction of SIRT6 when cells were senescent, compared with those young cells. Finally, SIRT6 markedly rescued senescence induced by p27. Our findings indicate that SIRT6 decreases p27 acetylation, leading to its degradation via ubiquitin-proteasome pathway and then delays cellular senescence.
Collapse
Affiliation(s)
- Ganye Zhao
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Hui Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Chenzhong Xu
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Pan Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Jun Chen
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Pengfeng Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Zhaomeng Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Yuanyuan Su
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Zhao Wang
- MOE Key Laboratory of Protein Sciences, Department of Pharmacology, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Limin Han
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| |
Collapse
|
218
|
Beckervordersandforth R. Mitochondrial Metabolism-Mediated Regulation of Adult Neurogenesis. Brain Plast 2017; 3:73-87. [PMID: 29765861 PMCID: PMC5928529 DOI: 10.3233/bpl-170044] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The life-long generation of new neurons from radial glia-like neural stem cells (NSCs) is achieved through a stereotypic developmental sequence that requires precise regulatory mechanisms to prevent exhaustion or uncontrolled growth of the stem cell pool. Cellular metabolism is the new kid on the block of adult neurogenesis research and the identity of stage-specific metabolic programs and their impact on neurogenesis turns out to be an emerging research topic in the field. Mitochondrial metabolism is best known for energy production but it contains a great deal more. Mitochondria are key players in a variety of cellular processes including ATP synthesis through functional coupling of the electron transport chain and oxidative phosphorylation, recycling of hydrogen carriers, biosynthesis of cellular building blocks, and generation of reactive oxygen species that can modulate signaling pathways in a redox-dependent fashion. In this review, I will discuss recent findings describing stage-specific modulations of mitochondrial metabolism within the adult NSC lineage, emphasizing its importance for NSC self-renewal, proliferation of neural stem and progenitor cells (NSPCs), cell fate decisions, and differentiation and maturation of newborn neurons. I will furthermore summarize the important role of mitochondrial dysfunction in tissue regeneration and ageing, suggesting it as a potential therapeutic target for regenerative medicine practice.
Collapse
Affiliation(s)
- Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| |
Collapse
|
219
|
Anamika, Khanna A, Acharjee P, Acharjee A, Trigun SK. Mitochondrial SIRT3 and neurodegenerative brain disorders. J Chem Neuroanat 2017; 95:43-53. [PMID: 29129747 DOI: 10.1016/j.jchemneu.2017.11.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/16/2017] [Accepted: 11/08/2017] [Indexed: 12/19/2022]
Abstract
Sirtuins are highly conserved NAD+ dependent class III histone deacetylases and catalyze deacetylation and ADP ribosylation of a number of non-histone proteins. Since, they require NAD+ for their activity, the cellular level of Sirtuins represents redox status of the cells and thereby serves as bona fide metabolic stress sensors. Out of seven homologues of Sirtuins identified in mammals, SIRT3, 4 & 5 have been found to be localized and active in mitochondria. During recent past, clusters of protein substrates for SIRT3 have been identified in mitochondria and thereby advocating SIRT3 as the main mitochondrial Sirtuin which could be involved in protecting stress induced mitochondrial integrity and energy metabolism. As mitochondrial dysfunction underlies the pathogenesis of almost all neurodegenerative diseases, a role of SIRT3 becomes an arguable speculation in such brain disorders. Some recent findings demonstrate that SIRT3 over expression could prevent neuronal derangements in certain in vivo and in vitro models of aging and neurodegenerative brain disorders like; Alzheimer's disease, Huntington's disease, stroke etc. Similarly, loss of SIRT3 has been found to accelerate neurodegeneration in the brain challenged with excitotoxicity. Therefore, it is argued that SIRT3 could be a relevant target to understand pathogenesis of neurodegenerative brain disorders. This review is an attempt to summarize recent findings on (1) the implication of SIRT3 in neurodegenerative brain disorders and (2) whether SIRT3 modulation could ameliorate neuropathologies in relevant models.
Collapse
Affiliation(s)
- Anamika
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Archita Khanna
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Arup Acharjee
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India.
| |
Collapse
|
220
|
Chini CCS, Tarragó MG, Chini EN. NAD and the aging process: Role in life, death and everything in between. Mol Cell Endocrinol 2017; 455:62-74. [PMID: 27825999 PMCID: PMC5419884 DOI: 10.1016/j.mce.2016.11.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/22/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022]
Abstract
Life as we know it cannot exist without the nucleotide nicotinamide adenine dinucleotide (NAD). From the simplest organism, such as bacteria, to the most complex multicellular organisms, NAD is a key cellular component. NAD is extremely abundant in most living cells and has traditionally been described to be a cofactor in electron transfer during oxidation-reduction reactions. In addition to participating in these reactions, NAD has also been shown to play a key role in cell signaling, regulating several pathways from intracellular calcium transients to the epigenetic status of chromatin. Thus, NAD is a molecule that provides an important link between signaling and metabolism, and serves as a key molecule in cellular metabolic sensoring pathways. Importantly, it has now been clearly demonstrated that cellular NAD levels decline during chronological aging. This decline appears to play a crucial role in the development of metabolic dysfunction and age-related diseases. In this review we will discuss the molecular mechanisms responsible for the decrease in NAD levels during aging. Since other reviews on this subject have been recently published, we will concentrate on presenting a critical appraisal of the current status of the literature and will highlight some controversial topics in the field. In particular, we will discuss the potential role of the NADase CD38 as a driver of age-related NAD decline.
Collapse
Affiliation(s)
- Claudia C S Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Mariana G Tarragó
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Eduardo N Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
221
|
Insights into the Dichotomous Regulation of SOD2 in Cancer. Antioxidants (Basel) 2017; 6:antiox6040086. [PMID: 29099803 PMCID: PMC5745496 DOI: 10.3390/antiox6040086] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
While loss of antioxidant expression and the resultant oxidant-dependent damage to cellular macromolecules is key to tumorigenesis, it has become evident that effective oxidant scavenging is conversely necessary for successful metastatic spread. This dichotomous role of antioxidant enzymes in cancer highlights their context-dependent regulation during different stages of tumor development. A prominent example of an antioxidant enzyme with such a dichotomous role and regulation is the mitochondria-localized manganese superoxide dismutase SOD2 (MnSOD). SOD2 has both tumor suppressive and promoting functions, which are primarily related to its role as a mitochondrial superoxide scavenger and H₂O₂ regulator. However, unlike true tumor suppressor- or onco-genes, the SOD2 gene is not frequently lost, or rarely mutated or amplified in cancer. This allows SOD2 to be either repressed or activated contingent on context-dependent stimuli, leading to its dichotomous function in cancer. Here, we describe some of the mechanisms that underlie SOD2 regulation in tumor cells. While much is known about the transcriptional regulation of the SOD2 gene, including downregulation by epigenetics and activation by stress response transcription factors, further research is required to understand the post-translational modifications that regulate SOD2 activity in cancer cells. Moreover, future work examining the spatio-temporal nature of SOD2 regulation in the context of changing tumor microenvironments is necessary to allows us to better design oxidant- or antioxidant-based therapeutic strategies that target the adaptable antioxidant repertoire of tumor cells.
Collapse
|
222
|
Alterations of sirtuins in mitochondrial cytochrome c-oxidase deficiency. PLoS One 2017; 12:e0186517. [PMID: 29059204 PMCID: PMC5653369 DOI: 10.1371/journal.pone.0186517] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 10/03/2017] [Indexed: 11/19/2022] Open
Abstract
Background Sirtuins are NAD+ dependent deacetylases, which regulate mitochondrial energy metabolism as well as cellular response to stress. The NAD/NADH-system plays a crucial role in oxidative phosphorylation linking sirtuins and the mitochondrial respiratory chain. Furthermore, sirtuins are able to directly deacetylate and activate different complexes of the respiratory chain. This prompted us to analyse sirtuin levels in skin fibroblasts from patients with cytochrome c-oxidase (COX) deficiency and to test the impact of different pharmaceutical activators of sirtuins (SRT1720, paeonol) to modulate sirtuins and possibly respiratory chain enzymes in patient cells in vitro. Methods We assayed intracellular levels of sirtuin 1 and the mitochondrial sirtuins SIRT3 and SIRT4 in human fibroblasts from patients with COX- deficiency. Furthermore, sirtuins were measured after inhibiting complex IV in healthy control fibroblasts by cyanide and after incubation with activators SRT1720 and paeonol. To determine the effect of sirtuin inhibition at the cellular level we measured total cellular acetylation (control and patient cells, with and without treatment) by Western blot. Results We observed a significant decrease in cellular levels of all three sirtuins at the activity, protein and transcriptional level (by 15% to 50%) in COX-deficient cells. Additionally, the intracellular concentration of NAD+ was reduced in patient cells. We mimicked the biochemical phenotype of COX- deficiency by incubating healthy fibroblasts with cyanide and observed reduced sirtuin levels. A pharmacological activation of sirtuins resulted in normalized sirtuin levels in patient cells. Hyper acetylation was also reversible after treatment with sirtuin activators. Pharmacological modulation of sirtuins resulted in altered respiratory chain complex activities. Conclusions We found inhibition of situins 1, 3 and 4 at activity, protein and transcriptional levels in fibroblasts from patient with COX-deficiency. Pharmacological activators were able to restore reduced sirtuin levels and thereby modulate respiratory chain activities.
Collapse
|
223
|
Chang TC, Hsu MF, Shih CY, Wu KK. 5-methoxytryptophan protects MSCs from stress induced premature senescence by upregulating FoxO3a and mTOR. Sci Rep 2017; 7:11133. [PMID: 28894133 PMCID: PMC5593915 DOI: 10.1038/s41598-017-11077-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022] Open
Abstract
5-methoxytryptophan (5-MTP) is a newly discovered tryptophan metabolite which controls stress-induced inflammatory signals. To determine whether 5-MTP protects against stress-induced mesenchymal stem cell (MSC) senescence, we incubated bone marrow-derived MSC (BM-MSC) in high-glucose medium or regular medium for 2 weeks followed by addiction of 5-MTP (10 μM) or vehicle for 48 h. 5-MTP reduced p16 and p21 expression, senescence-associated β-Gal (SA-β-Gal) and IL-6 secretion and increased BrdU incorporation. 5-MTP exerted a similar effect on BM-MSC senescence induced by a sublethal concentration of H2O2. 5-MTP enhanced FoxO3a expression and increased superoxide dismutase and catalase activities in HG BM-MSCs. Silencing of FoxO3a with siRNA abrogated 5-MTP-mediated reduction of SA-β-Gal and IL-6 secretion but not p21 or p16. Since mechanistic target of rapamycin (mTOR) is involved in cellular senescence, we determined whether 5-MTP influences mTOR expression. Our data reveal that mTOR protein level was depressed in HG-MSC which was rescued by 5-MTP. Rapamycin abrogated 5-MTP-mediated suppression of p16, p21, SA-β-Gal and IL-6 and rise of BrdU incorporation. Our findings suggest that 5-MTP protects MSCs against stress-induced senescence via FoxO3a and mTOR upregulation and has potential to improve cell expansion for cell therapy.
Collapse
Affiliation(s)
- Tzu-Ching Chang
- Metabolomic Medicine Research Center China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Min-Fen Hsu
- Metabolomic Medicine Research Center China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Yueh Shih
- Metabolomic Medicine Research Center China Medical University Hospital, Taichung, Taiwan
| | - Kenneth K Wu
- Metabolomic Medicine Research Center China Medical University Hospital, Taichung, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
224
|
Rupp M, Hagenbuchner J, Rass B, Fiegl H, Kiechl-Kohlendorfer U, Obexer P, Ausserlechner MJ. FOXO3-mediated chemo-protection in high-stage neuroblastoma depends on wild-type TP53 and SESN3. Oncogene 2017; 36:6190-6203. [PMID: 28869600 PMCID: PMC5671944 DOI: 10.1038/onc.2017.288] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/21/2017] [Accepted: 07/13/2017] [Indexed: 12/12/2022]
Abstract
Forkhead box O class transcription factors are homeostasis regulators that control cell death, longevity and therapy-resistance. In neuroblastoma (NB), nuclear FOXO3 correlates with stage M disease and poor prognosis. To analyze whether FOXO3 contributes to drug-resistance in this childhood cancer, we investigated how different high-stage-derived NB cells respond to the activation of an ectopic FOXO3 allele. We found endogenous FOXO3 mostly localized to the nucleus—upon activation of an ectopic, 4OHT-activated FOXO3(A3)ER fusion protein two of the cell lines underwent apoptosis, whereas in the others FOXO3-activation even increased survival during drug-treatment. In the latter cell type, FOXO3 did not induce the BH3-only protein BCL2L11/BIM due to impaired binding of FOXO3 to the BIM-promoter, but still activated other FOXO3 targets. It was shown before that FOXO3 and TP53 physically interact with each other at two different regions—the TP53-N-terminus binds to the FOXO3-DNA binding domain (DBD) and the FOXO3-C-terminus interacts with the TP53-DBD. Interestingly, cell lines that undergo FOXO3-induced cell death carry homozygous point mutations in the TP53-DBD near the structural hotspot-mutation-site R175H, which abrogated FOXO3–TP53 interaction. In contrast, in FOXO3-death-resistant cells no point mutations in the TP53-DBD were found—in these cells FOXO3–TP53 complexes are formed and FOXO3-binding to the BIM-promoter, but not the induction of the detoxifying protein SESN3, were prevented, which in turn increased chemo-protection in this type of high-stage-derived NB cells. Our combined data suggest that FOXO3 steps in as a death inducer in case of TP53-mutation, whereas functional TP53 alters FOXO3-target-promoter-recognition, which prevents death induction by FOXO3 and instead increases chemo-protection and survival of NB cells. This novel mechanism may explain the low incidence of TP53 mutation in high-stage NB at diagnosis and suggests FOXO3 as a therapeutic target for this childhood malignancy.
Collapse
Affiliation(s)
- M Rupp
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria.,Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - J Hagenbuchner
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria
| | - B Rass
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| | - H Fiegl
- Department of Obstetrics and Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | | | - P Obexer
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - M J Ausserlechner
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
225
|
Mariani S, Di Rocco G, Toietta G, Russo MA, Petrangeli E, Salvatori L. Sirtuins 1-7 expression in human adipose-derived stem cells from subcutaneous and visceral fat depots: influence of obesity and hypoxia. Endocrine 2017; 57:455-463. [PMID: 27844208 DOI: 10.1007/s12020-016-1170-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 11/03/2016] [Indexed: 01/14/2023]
Abstract
The sirtuin family comprises seven NAD+-dependent deacetylases which control the overall health of organisms through the regulation of pleiotropic metabolic pathways. Sirtuins are important modulators of adipose tissue metabolism and their expression is higher in lean than obese subjects. At present, the role of sirtuins in adipose-derived stem cells has not been investigated yet. Therefore, in this study, we evaluated the expression of the complete panel of sirtuins in adipose-derived stem cells isolated from both subcutaneous and visceral fat of non-obese and obese subjects. We aimed at investigating the influence of obesity on sirtuins' levels, their role in obesity-associated inflammation, and the relationship with the peroxisome proliferator-activated receptor delta, which also plays functions in adipose tissue metabolism. The mRNA levels in the four types of adipose-derived stem cells were evaluated by quantitative polymerase chain reaction, in untreated cells and also after 8 h of hypoxia exposure. Correlations among sirtuins' expression and clinical and molecular parameters were also analyzed. We found that sirtuin1-6 exhibited significant higher mRNA expression in visceral adipose-derived stem cells compared to subcutaneous adipose-derived stem cells of non-obese subjects. Sirtuin1-6 levels were markedly reduced in visceral adipose-derived stem cells of obese patients. Sirtuins' expression in visceral adipose-derived stem cells correlated negatively with body mass index and C-reactive protein and positively with peroxisome proliferator-activated receptor delta. Finally, only in the visceral adipose-derived stem cells of obese patients hypoxia-induced mRNA expression of all of the sirtuins. Our results highlight that sirtuins' levels in adipose-derived stem cells are consistent with protective effects against visceral obesity and inflammation, and suggest a transcriptional mechanism through which acute hypoxia up-regulates sirtuins in the visceral adipose-derived stem cells of obese patients.
Collapse
Affiliation(s)
- Stefania Mariani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Matteo A Russo
- Consorzio MEBIC, San Raffaele University, Laboratory of Molecular and Cellular Pathology, Rome, Italy
| | - Elisa Petrangeli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Luisa Salvatori
- Institute of Molecular Biology and Pathology, CNR, Rome, Italy.
| |
Collapse
|
226
|
Pande S, Kratasyuk VA, Medvedeva NN, Kolenchukova OA, Salmina AB. Nutritional biomarkers: Current view and future perspectives. Crit Rev Food Sci Nutr 2017; 58:3055-3069. [PMID: 28678523 DOI: 10.1080/10408398.2017.1350136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a poor relationship between nutrient intake and existing nutritional biomarkers due to variety of factors affecting their sensitivity and specificity. To explore the impact of nutrients at molecular level and devising a sensitive biomarker, proteomics is a central technology with sirtuins as one of the most promising nutritional biomarker. Sirtuins (seven mammalian sirtuins reported so far) have been reported to perform protein deacetylases and ADP-ribosyltransferases activity. It is distributed in different cellular compartments thereby controlling several metabolic processes. Sirtuins are oxidized nicotinamide adenine dinucleotide dependent, which implicates a direct effect of the metabolic state of the cell on its activity. Calorie restriction upregulates the mammalian sirtuin protein levels in variety of tissues and organs where it acts upon both histone and nonhistone substrates. Sirtuin senses nutrient availability and impacts gluconeogenesis, glycolysis, and insulin sensitivity. It deacetylates and inhibits the nuclear receptor that activates fat synthesis and adipogenesis in the body, leading to fat loss and bringing favorable cellular and health changes. Sirtuins mediates intracellular response that promotes cell survival, DNA damage repair thereby increasing the cell longitivity. The activation of sirtuins brings a wide spectrum of other health benefits and its activity levels are indicative of nutritional status as well as disease progression in cancer, inflammation, obesity, cardiovascular diseases, and viral infections. There are several foods that activate sirtuin activity and offer significant health benefits by their consumption.
Collapse
Affiliation(s)
- Shubhra Pande
- a Laboratory of Bioluminescent Biotechnologies, Department of Biophysics , Institute of Fundamental Biology and Biotechnology, Siberian Federal University , Krasnoyarsk , Russia.,b Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky , Krasnoyarsk , Russia
| | - Valentina A Kratasyuk
- a Laboratory of Bioluminescent Biotechnologies, Department of Biophysics , Institute of Fundamental Biology and Biotechnology, Siberian Federal University , Krasnoyarsk , Russia.,c Federal State Budgetary Scientific Institution "Institute of Biophysics, Siberian Branch of RAS" , Krasnoyarsk , Russia
| | - Nadezhda N Medvedeva
- b Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky , Krasnoyarsk , Russia
| | - Oxana A Kolenchukova
- a Laboratory of Bioluminescent Biotechnologies, Department of Biophysics , Institute of Fundamental Biology and Biotechnology, Siberian Federal University , Krasnoyarsk , Russia.,d Federal State Budgetary Scientific Institution "Scientific Research Institute of medical problems of the North" , Krasnoyarsk , Russia
| | - Alla B Salmina
- b Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky , Krasnoyarsk , Russia
| |
Collapse
|
227
|
Szegő ÉM, Gerhardt E, Outeiro TF. Sirtuin 2 enhances dopaminergic differentiation via the AKT/GSK-3β/β-catenin pathway. Neurobiol Aging 2017; 56:7-16. [DOI: 10.1016/j.neurobiolaging.2017.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 03/19/2017] [Accepted: 04/01/2017] [Indexed: 01/02/2023]
|
228
|
Mahalingaiah PKS, Ponnusamy L, Singh KP. Oxidative stress-induced epigenetic changes associated with malignant transformation of human kidney epithelial cells. Oncotarget 2017; 8:11127-11143. [PMID: 27655674 PMCID: PMC5355252 DOI: 10.18632/oncotarget.12091] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022] Open
Abstract
Renal Cell Carcinoma (RCC) in humans is positively influenced by oxidative stress status in kidneys. We recently reported that adaptive response to low level of chronic oxidative stress induces malignant transformation of immortalized human renal tubular epithelial cells. Epigenetic alterations in human RCC are well documented, but its role in oxidative stress-induced malignant transformation of kidney cells is not known. Therefore, the objective of this study was to evaluate the potential role of epigenetic changes in chronic oxidative stress-induced malignant transformation of HK-2, human renal tubular epithelial cells. The results revealed aberrant expression of epigenetic regulatory genes involved in DNA methylation (DNMT1, DNMT3a and MBD4) and histone modifications (HDAC1, HMT1 and HAT1) in HK-2 cells malignantly transformed by chronic oxidative stress. Additionally, both in vitro soft agar assay and in vivo nude mice study showing decreased tumorigenic potential of malignantly transformed HK-2 cells following treatment with DNA de-methylating agent 5-aza 2’ dC further confirmed the crucial role of DNA hypermethyaltion in oxidative stress-induced malignant transformation. Changes observed in global histone H3 acetylation (H3K9, H3K18, H3K27 and H3K14) and decrease in phospho-H2AX (Ser139) also suggest potential role of histone modifications in increased survival and malignant transformation of HK-2 cells by oxidative stress. In summary, the results of this study suggest that epigenetic reprogramming induced by low levels of oxidative stress act as driver for malignant transformation of kidney epithelial cells. Findings of this study are highly relevant in potential clinical application of epigenetic-based therapeutics for treatments of kidney cancers.
Collapse
Affiliation(s)
- Prathap Kumar S Mahalingaiah
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, USA
| | - Logeswari Ponnusamy
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, USA
| | - Kamaleshwar P Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
229
|
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
|
230
|
Garcia-Peterson LM, Wilking-Busch MJ, Ndiaye MA, Philippe CGA, Setaluri V, Ahmad N. Sirtuins in Skin and Skin Cancers. Skin Pharmacol Physiol 2017; 30:216-224. [PMID: 28704830 PMCID: PMC5568696 DOI: 10.1159/000477417] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022]
Abstract
The sirtuins are a family of proteins that comprise class III of the histone deacetylases. These NAD+-dependent proteins have been found to be intricately involved in a variety of important and skin-relevant cellular functions and processes, including aging, UV damage response, oxidative stress, and wound repair. In addition, recent research is unraveling the role of sirtuins in a variety of skin diseases, including melanoma and nonmelanoma skin cancers. In this review, we provide a discussion on the potential roles and implications of different sirtuins in skin-specific cellular processes, which may have relevance to skin health and skin diseases. Based on the available literature, the sirtuins appear to be important targets in the management of a variety of skin diseases from cosmetic (e.g., skin aging) to fatal conditions (e.g., melanoma).
Collapse
Affiliation(s)
| | | | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | | | - Vijayasaradhi Setaluri
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
- William S. Middleton VA Medical Center, Madison, Wisconsin, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
- William S. Middleton VA Medical Center, Madison, Wisconsin, USA
| |
Collapse
|
231
|
Guo Q, Li S, Xie Y, Zhang Q, Liu M, Xu Z, Sun H, Yang Y. The NAD +-dependent deacetylase, Bifidobacterium longum Sir2 in response to oxidative stress by deacetylating SigH (σ H) and FOXO3a in Bifidobacterium longum and HEK293T cell respectively. Free Radic Biol Med 2017; 108:929-939. [PMID: 28506746 DOI: 10.1016/j.freeradbiomed.2017.05.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 12/25/2022]
Abstract
Silent information regulator 2 (Sir2) enzymes which catalyze NAD+-dependent protein/histone deacetylation. The mammalian sirtuin family SIRT1, SIRT2, SIRT3 and SIRT6 can regulate oxidative stress. The probiotics (Bifidobacterium longum(B.longum) and Lactobacillus acidophilus(L. acidophilus)) have Sir2 gene family and have antioxidant activity in human body. it remains unknown whether probiotics Sir2 has a direct role in regulating oxidative stress. To this end, we knockout BL-sir2(sir2 B. longum) and LA-sir2(sir2 L.acidophilus) in low oxygen level. The antioxidant activities of two sir2 deficient strains was decreased, while when reintroduction of BL-sir2 and LA-sir2, the antioxidant activities were recoveried. In order to understand the regulation mechanism of probiotics Sir2 oxidation response. Then, we screened 65 acetylated protein, and found that SigH (σH) was a substrate of BL-Sir2. In addition, the acetylation level of σH decreased with the increase of BL-Sir2 level in B. longum. Thus, BL-Sir2 deacetylated σH in response to oxidative stress. Next, we transfected BL-Sir2 into H2O2-induced oxidative damage of 293T cells, BL-Sir2 increased the activity of manganese superoxide dismutase (MnSOD/SOD2) and catalase (CAT) and reduced reactive oxygen species(ROS). Then, we analyzed the differential gene by RNA sequencing and Gene ontology (GO) and found that BL-Sir2 regulated forkhead transcription factor (FOXO3a) mediated antioxidant genes in overexpressed BL-Sir2 HEK293T cells. Our study is the first to link probiotics Sir2 with oxidative stress and uncover the antioxidant mechanism of BL-Sir2 in B. longum itself and human body.
Collapse
Affiliation(s)
- Qing Guo
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Shiyu Li
- Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Yajie Xie
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Qian Zhang
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Mengge Liu
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhenrui Xu
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Hanxiao Sun
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Yan Yang
- Research Center of Agricultural and Sideline Products Processing, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| |
Collapse
|
232
|
Yang L, Ma X, Yuan C, He Y, Li L, Fang S, Xia W, He T, Qian S, Xu Z, Li G, Wang Z. Discovery of 2-((4,6-dimethylpyrimidin-2-yl)thio)- N -phenylacetamide derivatives as new potent and selective human sirtuin 2 inhibitors. Eur J Med Chem 2017; 134:230-241. [DOI: 10.1016/j.ejmech.2017.04.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 04/01/2017] [Accepted: 04/06/2017] [Indexed: 01/19/2023]
|
233
|
She DT, Jo DG, Arumugam TV. Emerging Roles of Sirtuins in Ischemic Stroke. Transl Stroke Res 2017; 8:10.1007/s12975-017-0544-4. [PMID: 28656393 DOI: 10.1007/s12975-017-0544-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is one of the leading causes of death worldwide. It is characterized by a sudden disruption of blood flow to the brain causing cell death and damage, which will lead to neurological impairments. In the current state, only one drug is approved to be used in clinical setting and new therapies that confer ischemic neuroprotection are desperately needed. Several targets and pathways have been indicated to be neuroprotective in ischemic stroke, among which the sirtuin family of nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases has emerged as important modulators of several processes in the normal physiology and pathological conditions such as stroke. Recent studies have identified some members of the sirtuin family are able to ameliorate the devastating consequences of ischemic stroke by conferring neuroprotection by means of reducing neuronal cell death, oxidative stress, and neuroinflammation whereas some sirtuins are found to be detrimental in the pathophysiology of ischemic stroke. This review summarizes implications of sirtuins in ischemic stroke and the experimental evidences that demonstrate the potential of sirtuin modulators as neuroprotective therapy for ischemic stroke.
Collapse
Affiliation(s)
- David T She
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Neurobiology/Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Neurobiology/Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
234
|
Chuang PY, Cai W, Li X, Fang L, Xu J, Yacoub R, He JC, Lee K. Reduction in podocyte SIRT1 accelerates kidney injury in aging mice. Am J Physiol Renal Physiol 2017; 313:F621-F628. [PMID: 28615249 PMCID: PMC5625108 DOI: 10.1152/ajprenal.00255.2017] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/09/2017] [Accepted: 06/11/2017] [Indexed: 11/22/2022] Open
Abstract
Both the incidence and prevalence of chronic kidney disease are increasing in the elderly population. Although aging is known to induce kidney injury, the underlying molecular mechanisms remain unclear. Sirtuin 1 (Sirt1), a longevity gene, is known to protect kidney cell injury from various cellular stresses. In previous studies, we showed that the podocyte-specific loss of Sirt1 aggravates diabetic kidney injury. However, the role of Sirt1 in aging-induced podocyte injury is not known. Therefore, in this study we sought to determine the effects of podocyte-specific reduction of Sirt1 in age-induced kidney injury. We employed the inducible podocyte-specific Sirt1 knockdown mice that express shRNA against Sirt1 (Pod-Sirt1RNAi) and control mice that express shRNA for luciferase (Pod-LuciRNAi). We found that reduction of podocyte Sirt1 led to aggravated aging-induced glomerulosclerosis and albuminuria. In addition, urinary level of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a marker of oxidative stress, was markedly increased in aged Pod-Sirt1RNAi mice compared with aged Pod-LuciRNAi mice. Although podocyte-specific markers decreased in aged mice compared with the young controls, the decrease was further exacerbated in aged Pod-Sirt1RNAi compared with Pod-LuciRNAi mice. Interestingly, expression of cellular senescence markers was significantly higher in the glomeruli of Pod-Sirt1RNAi mice than Pod-LuciRNAi mice, suggesting that cellular senescence may contribute to podocyte loss in aging kidneys. Finally, we confirmed that Pod-Sirt1RNAi glomeruli were associated with reduced activation of the transcription factors peroxisome proliferator-activated receptor (PPAR)-α coactivador-1 (PGC1α)/PPARγ, forkhead box O (FOXO)3, FOXO4, and p65 NF-κB, through SIRT1-mediated deacetylation. Together, our data suggest that SIRT1 may be a potential therapeutic target to treat patients with aging-related kidney disease.
Collapse
Affiliation(s)
- Peter Y Chuang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Weijing Cai
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Xuezhu Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Nephrology, Shanghai East Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Lu Fang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Nephrology, Shanghai East Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Jin Xu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rabi Yacoub
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Division of Nephrology, SUNY at Buffalo, Buffalo, New York; and
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; .,Renal Section, James J. Peters Veterans Affairs Medical Center, Bronx, New York
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
235
|
Sirtuin-2 Protects Neural Cells from Oxidative Stress and Is Elevated in Neurodegeneration. PARKINSONS DISEASE 2017. [PMID: 28634568 PMCID: PMC5467326 DOI: 10.1155/2017/2643587] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sirtuins are highly conserved lysine deacetylases involved in ageing, energy production, and lifespan extension. The mammalian SIRT2 has been implicated in Parkinson's disease (PD) where studies suggest SIRT2 promotes neurodegeneration. We therefore evaluated the effects of SIRT2 manipulation in toxin treated SH-SY5Y cells and determined the expression and activity of SIRT2 in postmortem brain tissue from patients with PD. SH-SY5Y viability in response to oxidative stress induced by diquat or rotenone was measured following SIRT2 overexpression or inhibition of deacetylase activity, along with α-synuclein aggregation. SIRT2 in human tissues was evaluated using Western blotting, immunohistochemistry, and fluorometric activity assays. In SH-SY5Y cells, elevated SIRT2 protected cells from rotenone or diquat induced cell death and enzymatic inhibition of SIRT2 enhanced cell death. SIRT2 protection was mediated, in part, through elevated SOD2 expression. SIRT2 reduced the formation of α-synuclein aggregates but showed minimal colocalisation with α-synuclein. In postmortem PD brain tissue, SIRT2 activity was elevated compared to controls but also elevated in other neurodegenerative disorders. Results from both in vitro work and brain tissue suggest that SIRT2 is necessary for protection against oxidative stress and higher SIRT2 activity in PD brain may be a compensatory mechanism to combat neuronal stress.
Collapse
|
236
|
Vojacek S, Beese K, Alhalabi Z, Swyter S, Bodtke A, Schulzke C, Jung M, Sippl W, Link A. Three-Component Aminoalkylations Yielding Dihydronaphthoxazine-Based Sirtuin Inhibitors: Scaffold Modification and Exploration of Space for Polar Side-Chains. Arch Pharm (Weinheim) 2017; 350. [DOI: 10.1002/ardp.201700097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/10/2017] [Accepted: 04/17/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Steffen Vojacek
- Institute of Pharmacy; University of Greifswald; Greifswald Germany
| | - Katja Beese
- Institute of Pharmacy; University of Greifswald; Greifswald Germany
| | - Zayan Alhalabi
- Institute of Pharmacy; Martin Luther University of Halle-Wittenberg; Halle/Saale Germany
| | - Sören Swyter
- Institute of Pharmaceutical Sciences; University of Freiburg; Freiburg Germany
| | - Anja Bodtke
- Institute of Pharmacy; University of Greifswald; Greifswald Germany
| | - Carola Schulzke
- Institute of Biochemistry; University of Greifswald; Greifswald Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences; University of Freiburg; Freiburg Germany
| | - Wolfgang Sippl
- Institute of Pharmacy; Martin Luther University of Halle-Wittenberg; Halle/Saale Germany
| | - Andreas Link
- Institute of Pharmacy; University of Greifswald; Greifswald Germany
| |
Collapse
|
237
|
Yang W, Gao F, Zhang P, Pang S, Cui Y, Liu L, Wei G, Yan B. Functional genetic variants within the SIRT2 gene promoter in acute myocardial infarction. PLoS One 2017; 12:e0176245. [PMID: 28445509 PMCID: PMC5406008 DOI: 10.1371/journal.pone.0176245] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/22/2017] [Indexed: 12/17/2022] Open
Abstract
Coronary artery disease (CAD), including acute myocardial infarction (AMI) is the complication of atherosclerosis. Recently, genome-wide association studies have identified a large number of CAD-related genetic variants. However, only 10% of CAD cases could be explained. Low frequent and rare genetic variants have been recently proposed to be main causes for CAD. SIRT2 is a member of sirtuin family, NAD(+)-dependent class III deacetylases. SIRT2 is involved in genomic stability, metabolism, inflammation, oxidative stress and autophagy, as well as in platelet function. Thus, we hypothesized that genetic variants in SIRT2 gene may contribute to AMI. In this study, SIRT2 gene promoter was analyzed in large cohorts of AMI patients (n = 375) and ethnic-matched controls (n = 377). Three novel heterozygous DSVs (g.38900888_91delTAAA, g.38900270A>G and g.38899853C>T) were identified in three AMI patients, but in none of controls. These DSVs significantly altered the transcriptional activity of the SIRT2 gene promoter (P<0.05) in both HEK-293 and H9c2 cells. Five novel heterozygous DSVS (g.38900562C>T, g.38900413A>C, g.38900030G>A, g.38899925A>C and g.38899852C>T) were only found in controls, which did not significantly affected SIRT2 gene promoter activity (P>0.05). In addition, four novel heterozygous DSVs and five SNPs were found in both AMI patients and control with similar frequencies (P>0.05), two SNPs of which were examined and did not affect SIRT2 gene promoter activity (P>0.05). Taken together, the DSVs identified in AMI patients may change SIRT2 level by affecting the transcriptional activity of SIRT2 gene promoter, contributing to the AMI development as a rare risk factor.
Collapse
Affiliation(s)
- Wentao Yang
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong, China
| | - Feng Gao
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong, China
| | - Pei Zhang
- College of Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Lixin Liu
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Guanghe Wei
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- * E-mail:
| |
Collapse
|
238
|
Cheng Q, Wu L, Tu R, Wu J, Kang W, Su T, Du R, Liu W. Mycoplasma fermentans deacetylase promotes mammalian cell stress tolerance. Microbiol Res 2017; 201:1-11. [PMID: 28602396 DOI: 10.1016/j.micres.2017.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 04/09/2017] [Accepted: 04/22/2017] [Indexed: 12/19/2022]
Abstract
Mycoplasma fermentans is a pathogenic bacterium that infects humans and has potential pathogenic roles in respiratory, genital and rheumatoid diseases. NAD+-dependent deacetylase is involved in a wide range of pathophysiological processes and our studies have demonstrated that expression of mycoplasmal deacetylase in mammalian cells inhibits proliferation but promotes anti-starvation stress tolerance. Furthermore, mycoplasmal deacetylase is involved in cellular anti-oxidation, which correlates with changes in the proapoptotic proteins BIK, p21 and BIM. Mycoplasmal deacetylase binds to and deacetylates the FOXO3 protein, similar with mammalian SIRT2, and affects expression of the FOXO3 target gene BIM, resulting in inhibition of cell proliferation. Mycoplasmal deacetylase also alters the performance of cells under drug stress. This study expands our understanding of the potential molecular and cellular mechanisms of interaction between mycoplasmas and mammalian cells.
Collapse
Affiliation(s)
- Qingzhou Cheng
- College of Health Sciences and Nursing, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Lijuan Wu
- College of Health Sciences and Nursing, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Rongfu Tu
- College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Jun Wu
- College of Health Sciences and Nursing, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Wenqian Kang
- College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Tong Su
- College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Runlei Du
- College of Life Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Wenbin Liu
- College of Health Sciences and Nursing, Wuhan Polytechnic University, Wuhan, Hubei, China; College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
239
|
Buechler N, Wang X, Yoza BK, McCall CE, Vachharajani V. Sirtuin 2 Regulates Microvascular Inflammation during Sepsis. J Immunol Res 2017; 2017:2648946. [PMID: 28503576 PMCID: PMC5414591 DOI: 10.1155/2017/2648946] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/23/2017] [Accepted: 03/05/2017] [Indexed: 12/21/2022] Open
Abstract
Objective. Sepsis and septic shock, the leading causes of death in noncoronary intensive care units, kill more than 200,000/year in the US alone. Circulating cell-endothelial cell interactions are the rate determining factor in sepsis inflammation. Sirtuin, a seven-member family of proteins (SIRT1-7), epigenetically controls inflammation. We have studied the roles of SIRTs 1, 3, and 6 in sepsis previously. In this project, we studied the role of SIRT2 on sepsis-related inflammation. Methods. Sepsis was induced in C57Bl/6 (WT), SIRT2 knockout (SIRT2KO), and SIRT2 overexpressing (SIRT2KI) mice by cecal ligation and puncture (CLP). We studied leukocyte/platelet adhesion using intravital microscopy and E-selectin/ICAM-1 adhesion molecule expression in the small intestine with immunohistochemistry (IHC) six hours post-CLP/sham surgery. We also studied 7-day survival rates in WT, SIRT2KO, and SIRT2KI sepsis mice. Results. Compared to WT mice, SIRT2KO mice show exaggeration while SIRT2KI mice show attenuation of cellular adhesion with sepsis in the small intestine. We also show that the small intestinal E-selectin and ICAM-1 expressions increased in SIRT2KO and decreased in SIRT2KI mice versus those in WT sepsis mice. We show that the 7-day survival rate is decreased in SIRT2KO and increased in SIRT2KI sepsis mice. Conclusion. SIRT2 modulates microvascular inflammation in sepsis and affects survival.
Collapse
Affiliation(s)
- Nancy Buechler
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Xianfeng Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barbara K. Yoza
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Charles E. McCall
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Vidula Vachharajani
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
240
|
Moniot S, Forgione M, Lucidi A, Hailu GS, Nebbioso A, Carafa V, Baratta F, Altucci L, Giacché N, Passeri D, Pellicciari R, Mai A, Steegborn C, Rotili D. Development of 1,2,4-Oxadiazoles as Potent and Selective Inhibitors of the Human Deacetylase Sirtuin 2: Structure–Activity Relationship, X-ray Crystal Structure, and Anticancer Activity. J Med Chem 2017; 60:2344-2360. [DOI: 10.1021/acs.jmedchem.6b01609] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sébastien Moniot
- Department
of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Mariantonietta Forgione
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
- Center
for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Alessia Lucidi
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| | - Gebremedhin S. Hailu
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| | - Angela Nebbioso
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Vincenzo Carafa
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Francesca Baratta
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Lucia Altucci
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Nicola Giacché
- TES Pharma S.r.l., Via P. Togliatti 20, 06073 Corciano, Perugia, Italy
| | - Daniela Passeri
- TES Pharma S.r.l., Via P. Togliatti 20, 06073 Corciano, Perugia, Italy
| | | | - Antonello Mai
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| | - Clemens Steegborn
- Department
of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Dante Rotili
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| |
Collapse
|
241
|
Phosphorylation and acetylation modifications of FOXO3a: Independently or synergistically? Oncol Lett 2017; 13:2867-2872. [PMID: 28521392 PMCID: PMC5431355 DOI: 10.3892/ol.2017.5851] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/26/2016] [Indexed: 12/25/2022] Open
Abstract
Forkhead box class O 3a (FOXO3a) is a transcription factor that has emerged as being a tumor suppressor and longevity factor. The precise regulation of FOXO3a transactivation of target genes is achieved via post-translational modifications (PTMs) and specific protein-protein interactions. The multiple types of PTMs that FOXO3a undergoes, including phosphorylation, acetylation, methylation and ubiquitination, serve important roles in directing its subcellular localization and transcription activity, which are central to the integration of insulin/growth factor signaling and oxidative/nutrient stress signaling. The present review summarizes the modifications of FOXO3a that occur via phosphorylation and acetylation. In addition, the synergistic effect of multiple phosphorylations on FOXO3a and the crosstalk between phosphorylation and acetylation in the regulation of FOXO3a are discussed. These discussions may highlight potential strategies for the prevention of cancer and aging.
Collapse
|
242
|
Grabowska W, Sikora E, Bielak-Zmijewska A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology 2017; 18:447-476. [PMID: 28258519 PMCID: PMC5514220 DOI: 10.1007/s10522-017-9685-9] [Citation(s) in RCA: 314] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 02/21/2017] [Indexed: 12/17/2022]
Abstract
Ageing is a plastic process and can be successfully modulated by some biomedical approaches or pharmaceutics. In this manner it is possible to delay or even prevent some age-related pathologies. There are some defined interventions, which give promising results in animal models or even in human studies, resulting in lifespan elongation or healthspan improvement. One of the most promising targets for anti-ageing approaches are proteins belonging to the sirtuin family. Sirtuins were originally discovered as transcription repressors in yeast, however, nowadays they are known to occur in bacteria and eukaryotes (including mammals). In humans the family consists of seven members (SIRT1-7) that possess either mono-ADP ribosyltransferase or deacetylase activity. It is believed that sirtuins play key role during cell response to a variety of stresses, such as oxidative or genotoxic stress and are crucial for cell metabolism. Although some data put in question direct involvement of sirtuins in extending human lifespan, it was documented that proper lifestyle including physical activity and diet can influence healthspan via increasing the level of sirtuins. The search for an activator of sirtuins is one of the most extensive and robust topic of research. Some hopes are put on natural compounds, including curcumin. In this review we summarize the involvement and usefulness of sirtuins in anti-ageing interventions and discuss the potential role of curcumin in sirtuins regulation.
Collapse
Affiliation(s)
- Wioleta Grabowska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Anna Bielak-Zmijewska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland.
| |
Collapse
|
243
|
Gribble KE, Mark Welch DB. Genome-wide transcriptomics of aging in the rotifer Brachionus manjavacas, an emerging model system. BMC Genomics 2017; 18:217. [PMID: 28249563 PMCID: PMC5333405 DOI: 10.1186/s12864-017-3540-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/02/2017] [Indexed: 12/22/2022] Open
Abstract
Background Understanding gene expression changes over lifespan in diverse animal species will lead to insights to conserved processes in the biology of aging and allow development of interventions to improve health. Rotifers are small aquatic invertebrates that have been used in aging studies for nearly 100 years and are now re-emerging as a modern model system. To provide a baseline to evaluate genetic responses to interventions that change health throughout lifespan and a framework for new hypotheses about the molecular genetic mechanisms of aging, we examined the transcriptome of an asexual female lineage of the rotifer Brachionus manjavacas at five life stages: eggs, neonates, and early-, late-, and post-reproductive adults. Results There are widespread shifts in gene expression over the lifespan of B. manjavacas; the largest change occurs between neonates and early reproductive adults and is characterized by down-regulation of developmental genes and up-regulation of genes involved in reproduction. The expression profile of post-reproductive adults was distinct from that of other life stages. While few genes were significantly differentially expressed in the late- to post-reproductive transition, gene set enrichment analysis revealed multiple down-regulated pathways in metabolism, maintenance and repair, and proteostasis, united by genes involved in mitochondrial function and oxidative phosphorylation. Conclusions This study provides the first examination of changes in gene expression over lifespan in rotifers. We detected differential expression of many genes with human orthologs that are absent in Drosophila and C. elegans, highlighting the potential of the rotifer model in aging studies. Our findings suggest that small but coordinated changes in expression of many genes in pathways that integrate diverse functions drive the aging process. The observation of simultaneous declines in expression of genes in multiple pathways may have consequences for health and longevity not detected by single- or multi-gene knockdown in otherwise healthy animals. Investigation of subtle but genome-wide change in these pathways during aging is an important area for future study. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3540-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kristin E Gribble
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| | - David B Mark Welch
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, 02543, USA.
| |
Collapse
|
244
|
Ansari A, Rahman MS, Saha SK, Saikot FK, Deep A, Kim KH. Function of the SIRT3 mitochondrial deacetylase in cellular physiology, cancer, and neurodegenerative disease. Aging Cell 2017; 16:4-16. [PMID: 27686535 PMCID: PMC5242307 DOI: 10.1111/acel.12538] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2016] [Indexed: 12/11/2022] Open
Abstract
In mammals, seven members of the sirtuin protein family known as class III histone deacetylase have been identified for their characteristic features. These distinguished characteristics include the tissues where they are distributed or located, enzymatic activities, molecular functions, and involvement in diseases. Among the sirtuin members, SIRT3 has received much attention for its role in cancer genetics, aging, neurodegenerative disease, and stress resistance. SIRT3 controls energy demand during stress conditions such as fasting and exercise as well as metabolism through the deacetylation and acetylation of mitochondrial enzymes. SIRT3 is well known for its ability to eliminate reactive oxygen species and to prevent the development of cancerous cells or apoptosis. This review article provides a comprehensive review on numerous (noteworthy) molecular functions of SIRT3 and its effect on cancer cells and various diseases including Huntington's disease, amyotrophic lateral sclerosis, and Alzheimer's disease.
Collapse
Affiliation(s)
- Aneesa Ansari
- Department of Genetic Engineering and Biotechnology; Jessore University of Science and Technology; Jessore 7408 Bangladesh
| | - Md. Shahedur Rahman
- Department of Genetic Engineering and Biotechnology; Jessore University of Science and Technology; Jessore 7408 Bangladesh
| | - Subbroto K. Saha
- Department of Stem Cell and Regenerative Biology; Konkuk University; 120 Neungdong-Ro Seoul 05029 Korea
| | - Forhad K. Saikot
- Department of Genetic Engineering and Biotechnology; Jessore University of Science and Technology; Jessore 7408 Bangladesh
| | - Akash Deep
- Central Scientific Instruments Organisation (CSIR-CSIO); Sector 30 C Chandigarh 160030 India
| | - Ki-Hyun Kim
- Department of Civil & Environmental Engineering; Hanyang University; 222 Wangsimni-Ro Seoul 04763 Korea
| |
Collapse
|
245
|
Sirtuins Expression and Their Role in Retinal Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3187594. [PMID: 28197299 PMCID: PMC5288547 DOI: 10.1155/2017/3187594] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/13/2016] [Indexed: 01/28/2023]
Abstract
Sirtuins have received considerable attention since the discovery that silent information regulator 2 (Sir2) extends the lifespan of yeast. Sir2, a nicotinamide adenine dinucleotide- (NAD-) dependent histone deacetylase, serves as both a transcriptional effector and energy sensor. Oxidative stress and apoptosis are implicated in the pathogenesis of neurodegenerative eye diseases. Sirtuins confer protection against oxidative stress and retinal degeneration. In mammals, the sirtuin (SIRT) family consists of seven proteins (SIRT1–SIRT7). These vary in tissue specificity, subcellular localization, and enzymatic activity and targets. In this review, we present the current knowledge of the sirtuin family and discuss their structure, cellular location, and biological function with a primary focus on their role in different neuroophthalmic diseases including glaucoma, optic neuritis, and age-related macular degeneration. The potential role of certain therapeutic targets is also described.
Collapse
|
246
|
Schiedel M, Robaa D, Rumpf T, Sippl W, Jung M. The Current State of NAD + -Dependent Histone Deacetylases (Sirtuins) as Novel Therapeutic Targets. Med Res Rev 2017; 38:147-200. [PMID: 28094444 DOI: 10.1002/med.21436] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 12/19/2022]
Abstract
Sirtuins are NAD+ -dependent protein deacylases that cleave off acetyl, as well as other acyl groups, from the ε-amino group of lysines in histones and other substrate proteins. Seven sirtuin isotypes (Sirt1-7) have been identified in mammalian cells. As sirtuins are involved in the regulation of various physiological processes such as cell survival, cell cycle progression, apoptosis, DNA repair, cell metabolism, and caloric restriction, a dysregulation of their enzymatic activity has been associated with the pathogenesis of neoplastic, metabolic, infectious, and neurodegenerative diseases. Thus, sirtuins are promising targets for pharmaceutical intervention. Growing interest in a modulation of sirtuin activity has prompted the discovery of several small molecules, able to inhibit or activate certain sirtuin isotypes. Herein, we give an update to our previous review on the topic in this journal (Schemies, 2010), focusing on recent developments in sirtuin biology, sirtuin modulators, and their potential as novel therapeutic agents.
Collapse
Affiliation(s)
- Matthias Schiedel
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Dina Robaa
- Department of Pharmaceutical Chemistry, Martin-Luther Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Tobias Rumpf
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Wolfgang Sippl
- Department of Pharmaceutical Chemistry, Martin-Luther Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
247
|
Carnevale I, Pellegrini L, D'Aquila P, Saladini S, Lococo E, Polletta L, Vernucci E, Foglio E, Coppola S, Sansone L, Passarino G, Bellizzi D, Russo MA, Fini M, Tafani M. SIRT1-SIRT3 Axis Regulates Cellular Response to Oxidative Stress and Etoposide. J Cell Physiol 2017; 232:1835-1844. [PMID: 27925196 DOI: 10.1002/jcp.25711] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/29/2016] [Indexed: 02/01/2023]
Abstract
Sirtuins are conserved NAD+ -dependent deacylases. SIRT1 is a nuclear and cytoplasmic sirtuin involved in the control of histones a transcription factors function. SIRT3 is a mitochondrial protein, which regulates mitochondrial function. Although, both SIRT1 and SIRT3 have been implicated in resistance to cellular stress, the link between these two sirtuins has not been studied so far. Here we aimed to unravel: i) the role of SIRT1-SIRT3 axis for cellular response to oxidative stress and DNA damage; ii) how mammalian cells modulate such SIRT1-SIRT3 axis and which mechanisms are involved. Therefore, we analyzed the response to different stress stimuli in WT or SIRT1-silenced cell lines. Our results demonstrate that SIRT1-silenced cells are more resistant to H2 O2 and etoposide treatment showing decreased ROS accumulation, γ-H2AX phosphorylation, caspase-3 activation and PARP cleavage. Interestingly, we observed that SIRT1-silenced cells show an increased SIRT3 expression. To explore such a connection, we carried out luciferase assays on SIRT3 promoter demonstrating that SIRT1-silencing increases SIRT3 promoter activity and that such an effect depends on the presence of SP1 and ZF5 recognition sequences on SIRT3 promoter. Afterwards, we performed co-immunoprecipitation assays demonstrating that SIRT1 binds and deacetylates the transcription inhibitor ZF5 and that there is a decreased interaction between SP1 and ZF5 in SIRT1-silenced cells. Therefore, we speculate that acetylated ZF5 cannot bind and sequester SP1 that is free, then, to increase SIRT3 transcription. In conclusion, we demonstrate that cells with low SIRT1 levels can maintain their resistance and survival by increasing SIRT3 expression. J. Cell. Physiol. 232: 1835-1844, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ilaria Carnevale
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - Laura Pellegrini
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico (CCP), Milan, Italy
| | - Patrizia D'Aquila
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Serena Saladini
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - Emanuela Lococo
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - Lucia Polletta
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - Enza Vernucci
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - Eleonora Foglio
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy
| | - Stefano Coppola
- Physics of Life Processes, Kammerlingh Onnes-Huygens Laboratory, Leiden University, Leiden, The Netherlands
| | - Luigi Sansone
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, Rome, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Dina Bellizzi
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | | | - Massimo Fini
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, University of Rome, Sapienza, Rome, Italy.,Department of Cellular and Molecular Pathology, IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
248
|
Correia M, Perestrelo T, Rodrigues AS, Ribeiro MF, Pereira SL, Sousa MI, Ramalho-Santos J. Sirtuins in metabolism, stemness and differentiation. Biochim Biophys Acta Gen Subj 2017; 1861:3444-3455. [DOI: 10.1016/j.bbagen.2016.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/16/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
|
249
|
Shin J, Yang SJ, Lim Y. Gamma-tocopherol supplementation ameliorated hyper-inflammatory response during the early cutaneous wound healing in alloxan-induced diabetic mice. Exp Biol Med (Maywood) 2016; 242:505-515. [PMID: 28211759 DOI: 10.1177/1535370216683836] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Delayed wound healing is one of the major diabetic complications. During wound healing process, the early inflammatory stage is important for better prognosis. One of antioxidant nutrient, gamma-tocopherol (GT) is considered to regulate inflammatory conditions. This study investigated the effect of GT supplementation on mechanism associated with inflammation, oxidative stress, and apoptosis during early cutaneous wound healing in diabetic mice. Diabetes was induced by alloxan injection in ICR mice. All mice were divided into three groups: non-diabetic control mice (CON), diabetic control mice (DMC), and diabetic mice supplemented with GT (GT). After two weeks of GT supplementation, excisional wounds were made by biopsy punches (4 mm). Diabetic mice showed increases in fasting blood glucose (FBG) level, hyper-inflammatory response, oxidative stress, and delayed wound closure rate compared to non-diabetic mice. However, GT supplementation reduced FBG level and accelerated wound closure rate by regulation of inflammatory response-related proteins such as nuclear factor kappa B, interleukin-1β, tumor necrosis factor-α, and c-reactive protein, and oxidative stress-related markers including nuclear factor (erythroid derived 2)-like 2, NAD(P)H dehydrogenase quinone1, heme oxygenase-1, manganese superoxide dismutase, catalase and glutathione peroxidase and apoptosis-related markers such as sirtuin-1, peroxisome proliferator-activated receptor gamma coactivator 1- α, and p53 in diabetic mice. Taken together, GT would be a potential therapeutic to prevent diabetes-induced delayed wound healing by regulation of inflammatory response, apoptosis, and oxidative stress. Impact statement Gamma tocopherol has shown ameliorative effect on diabetic wound healing by regulation of inflammation, oxidative stress, and apoptosis demonstrated by nuclear factor kappa B, nuclear factor (erythroid derived 2)-like 2, and sirtuin-1.
Collapse
Affiliation(s)
- Jihyun Shin
- 1 Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Korea
| | - Soo Jin Yang
- 2 Department of Food and Nutrition, Seoul Women's University, Seoul 01797, Korea
| | - Yunsook Lim
- 1 Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
250
|
Xie XQ, Zhang P, Tian B, Chen XQ. Downregulation of NAD-Dependent Deacetylase SIRT2 Protects Mouse Brain Against Ischemic Stroke. Mol Neurobiol 2016; 54:7251-7261. [DOI: 10.1007/s12035-016-0173-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/28/2016] [Indexed: 12/20/2022]
|