1
|
Bonomi RE, Riordan W, Gelovani JG. The Structures, Functions, and Roles of Class III HDACs (Sirtuins) in Neuropsychiatric Diseases. Cells 2024; 13:1644. [PMID: 39404407 PMCID: PMC11476333 DOI: 10.3390/cells13191644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Over the past two decades, epigenetic regulation has become a rapidly growing and influential field in biology and medicine. One key mechanism involves the acetylation and deacetylation of lysine residues on histone core proteins and other critical proteins that regulate gene expression and cellular signaling. Although histone deacetylases (HDACs) have received significant attention, the roles of individual HDAC isoforms in the pathogenesis of psychiatric diseases still require further research. This is particularly true with regard to the sirtuins, class III HDACs. Sirtuins have unique functional activity and significant roles in normal neurophysiology, as well as in the mechanisms of addiction, mood disorders, and other neuropsychiatric abnormalities. This review aims to elucidate the differences in catalytic structure and function of the seven sirtuins as they relate to psychiatry.
Collapse
Affiliation(s)
- Robin E. Bonomi
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA;
| | - William Riordan
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA;
| | - Juri G. Gelovani
- College of Medicine and Health Sciences, Office of the Provost, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Radiology, Division of Nuclear Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
2
|
A S, Sudhakar M, Nair AS, Kamalamma S. Identification and molecular modeling of novel endogenous activator proteins of Sirt-1: an in silico study. J Biomol Struct Dyn 2023; 41:8276-8291. [PMID: 36229233 DOI: 10.1080/07391102.2022.2132294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/28/2022] [Indexed: 10/17/2022]
Abstract
Sirt-1 is one of the most extensively studied mammalian Sirtuins that deacetylates histones and several non-histone proteins critical to cellular homeostasis. As a key sensor of cellular metabolism, it is regulated at multiple levels including transcriptional and post translational levels. As an allosteric enzyme, its activity is also modulated by ligands and certain endogenous proteins. The present study is an in silico approach to identify novel Sirt-1 binding proteins. Bioinformatic search for similarity in sequence, structure, and topology of binding region to Lamin-A, a known activator of Sirt-1, identified three proteins viz. Epididymis secretory sperm binding protein (ESSBP), xylosyltransferase 1 (XT-1), and Adenylyl cyclase 9 (ADCY-9). Molecular docking studies revealed binding of ESSBP and ADCY-9 to the N-terminal region of Sirt-1 while XT-1 docks at both N-terminal and C-terminal region of Sirt-1 with Z-Dock score better than Lamin-A; XT-1 and ADCY-9 showed better Z-Rank score as well. MD simulation studies for extended time followed by MM-PBSA analysis showed that the Sirt-1-protein complexes were stable with favourable binding energy and minimal change in RMSD relating to backbone structure and RMSF relating to residue fluctuations. Further, H-bond analysis showed only minimal changes in H bonding interactions. Docking of these proteins to Sirt-1 through interaction with several residues particularly to its N-terminal region spanning 1-243 residues, in a manner similar to the docking of the activator Lamin-A and different from the inhibitor DLBC-1 binding site, suggests that these proteins may also positively modulate Sirt-1 activity. Further experimental data would be required to validate the computational prediction and to understand its physiological role.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shanitha A
- Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, India
| | - Manu Sudhakar
- Amrita School of Medicine, Amrita Institute of Medical Sciences and Research center, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Achuthsankar S Nair
- Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, India
| | - Saja Kamalamma
- Department of Biochemistry, University of Kerala, Trivandrum, India
| |
Collapse
|
3
|
Hur Y, Huynh J, Leong E, Dosanjh R, Charvat AF, Vu MH, Alam Z, Lee YT, Cabreros CC, Carroll EC, Hura GL, Wang N. The differing effects of a dual acting regulator on SIRT1. Front Mol Biosci 2023; 10:1260489. [PMID: 37711385 PMCID: PMC10499324 DOI: 10.3389/fmolb.2023.1260489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/16/2023] Open
Abstract
SIRT1 is an NAD+-dependent protein deacetylase that has been shown to play a significant role in many biological pathways, such as insulin secretion, tumor formation, lipid metabolism, and neurodegeneration. There is great interest in understanding the regulation of SIRT1 to better understand SIRT1-related diseases and to better design therapeutic approaches that target SIRT1. There are many known protein and small molecule activators and inhibitors of SIRT1. One well-studied SIRT1 regulator, resveratrol, has historically been regarded as a SIRT1 activator, however, recent studies have shown that it can also act as an inhibitor depending on the identity of the peptide substrate. The inhibitory nature of resveratrol has yet to be studied in detail. Understanding the mechanism behind this dual behavior is crucial for assessing the potential side effects of STAC-based therapeutics. Here, we investigate the detailed mechanism of substrate-dependent SIRT1 regulation by resveratrol. We demonstrate that resveratrol alters the substrate recognition of SIRT1 by affecting the K M values without significantly impacting the catalytic rate (k cat). Furthermore, resveratrol destabilizes SIRT1 and extends its conformation, but the conformational changes differ between the activation and inhibition scenarios. We propose that resveratrol renders SIRT1 more flexible in the activation scenario, leading to increased activity, while in the inhibition scenario, it unravels the SIRT1 structure, compromising substrate recognition. Our findings highlight the importance of substrate identity in resveratrol-mediated SIRT1 regulation and provide insights into the allosteric control of SIRT1. This knowledge can guide the development of targeted therapeutics for diseases associated with dysregulated SIRT1 activity.
Collapse
Affiliation(s)
- Yujin Hur
- Department of Chemistry, San José State University, San José, CA, United States
| | - Johnson Huynh
- Department of Chemistry, San José State University, San José, CA, United States
| | - Emily Leong
- Department of Chemistry, San José State University, San José, CA, United States
| | - Reena Dosanjh
- Department of Chemistry, San José State University, San José, CA, United States
| | - Annemarie F. Charvat
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, United States
| | - My H. Vu
- Department of Chemistry, San José State University, San José, CA, United States
| | - Zain Alam
- Department of Chemistry, San José State University, San José, CA, United States
| | - Yue Tong Lee
- Department of Chemistry, San José State University, San José, CA, United States
| | | | - Emma C. Carroll
- Department of Chemistry, San José State University, San José, CA, United States
| | - Greg L. Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Ningkun Wang
- Department of Chemistry, San José State University, San José, CA, United States
| |
Collapse
|
4
|
Tang H, Wen J, Qin T, Chen Y, Huang J, Yang Q, Jiang P, Wang L, Zhao Y, Yang Q. New insights into Sirt1: potential therapeutic targets for the treatment of cerebral ischemic stroke. Front Cell Neurosci 2023; 17:1228761. [PMID: 37622049 PMCID: PMC10445043 DOI: 10.3389/fncel.2023.1228761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
Ischemic stroke is one of the main causes of mortality and disability worldwide. However, the majority of patients are currently unable to benefit from intravenous thrombolysis or intravascular mechanical thrombectomy due to the limited treatment windows and serious complications. Silent mating type information regulation 2 homolog 1 (Sirt1), a nicotine adenine dinucleotide-dependent enzyme, has emerged as a potential therapeutic target for ischemic stroke due to its ability to maintain brain homeostasis and possess neuroprotective properties in a variety of pathological conditions for the central nervous system. Animal and clinical studies have shown that activation of Sirt1 can lessen neurological deficits and reduce the infarcted volume, offering promise for the treatment of ischemic stroke. In this review, we summarized the direct evidence and related mechanisms of Sirt1 providing neuroprotection against cerebral ischemic stroke. Firstly, we introduced the protein structure, catalytic mechanism and specific location of Sirt1 in the central nervous system. Secondly, we list the activators and inhibitors of Sirt1, which are primarily divided into three categories: natural, synthetic and physiological. Finally, we reviewed the neuroprotective effects of Sirt1 in ischemic stroke and discussed the specific mechanisms, including reducing neurological deficits by inhibiting various programmed cell death such as pyroptosis, necroptosis, ferroptosis, and cuproptosis in the acute phase, as well as enhancing neurological repair by promoting angiogenesis and neurogenesis in the later stage. Our review aims to contribute to a deeper understanding of the critical role of Sirt1 in cerebral ischemic stroke and to offer novel therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Hao Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Wen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Qin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiagui Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghuan Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peiran Jiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Medoro A, Jafar TH, Ali S, Trung TT, Sorrenti V, Intrieri M, Scapagnini G, Davinelli S. In silico evaluation of geroprotective phytochemicals as potential sirtuin 1 interactors. Biomed Pharmacother 2023; 161:114425. [PMID: 36812712 DOI: 10.1016/j.biopha.2023.114425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Sirtuin 1 (SIRT1) belongs to the histone deacetylase enzyme family and its activity regulates various signaling networks associated with aging. SIRT1 is widely involved in a large number of biological processes, including senescence, autophagy, inflammation, and oxidative stress. In addition, SIRT1 activation may improve lifespan and health in numerous experimental models. Therefore, SIRT1 targeting is a potential strategy to delay or reverse aging and age-related diseases. Although SIRT1 is activated by a wide array of small molecules, only a limited number of phytochemicals that directly interact with SIRT1 have been identified. Using the Geroprotectors.org database and a literature search, the aim of this study was to identify geroprotective phytochemicals that might interact with SIRT1. We performed molecular docking, density functional theory studies, molecular dynamic simulations (MDS), and absorption, distribution, metabolism, excretion, and toxicity (ADMET) prediction to screen potential candidates against SIRT1. After the initial screening of 70 phytochemicals, crocin, celastrol, hesperidin, taxifolin, vitexin, and quercetin had significant binding affinity scores. These six compounds established multiple hydrogen-bonding and hydrophobic interactions with SIRT1 and showed good drug-likeness and ADMET properties. In particular, crocin was further analyzed using MDS to study its complex with SIRT1 during simulation. Crocin has a high reactivity to SIRT1 and can form a stable complex with it, showing a good ability to fit into the binding pocket. Although further investigations are required, our results suggest that these geroprotective phytochemicals, especially crocin, are novel interacting partners of SIRT1.
Collapse
Affiliation(s)
- Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Tassadaq Hussain Jafar
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Sawan Ali
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Truong Tan Trung
- Laboratory of Computation and Nanoscience, Dong Nai Technology University, Dong Nai, Vietnam
| | - Vincenzo Sorrenti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Mariano Intrieri
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy.
| | - Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| |
Collapse
|
6
|
Lee YH, Kim SJ, Surh YJ. Role of Post-translational Modification of Silent Mating Type Information Regulator 2 Homolog 1 in Cancer and Other Disorders. J Cancer Prev 2022; 27:157-169. [PMID: 36258719 PMCID: PMC9537581 DOI: 10.15430/jcp.2022.27.3.157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022] Open
Abstract
Silent mating type information regulator 2 homolog 1 (SIRT1), an NAD+-dependent histone/protein deacetylase, has multifarious physiological roles in development, metabolic regulation, and stress response. Thus, its abnormal expression or malfunction is implicated in pathogenesis of various diseases. SIRT1 undergoes post-translational modifications, including phosphorylation, oxidation/reduction, carbonylation, nitrosylation, glycosylation, ubiquitination/deubiquitination, SUMOylation etc. which can modulate its catalytic activity, stability, subcellular localization, and also binding affinity for substrate proteins. This short review highlights the regulation of SIRT1 post-translational modifications and their pathophysiologic implications.
Collapse
Affiliation(s)
- Yeon-Hwa Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul, Korea
| | - Su-Jung Kim
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul, Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul, Korea,Cancer Research Institute, Seoul National University, Seoul, Korea,Correspondence to Young-Joon Surh, E-mail: , https://orcid.org/0000-0001-8310-1795
| |
Collapse
|
7
|
Abbotto E, Scarano N, Piacente F, Millo E, Cichero E, Bruzzone S. Virtual Screening in the Identification of Sirtuins’ Activity Modulators. Molecules 2022; 27:molecules27175641. [PMID: 36080416 PMCID: PMC9457788 DOI: 10.3390/molecules27175641] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Sirtuins are NAD+-dependent deac(et)ylases with different subcellular localization. The sirtuins’ family is composed of seven members, named SIRT-1 to SIRT-7. Their substrates include histones and also an increasing number of different proteins. Sirtuins regulate a wide range of different processes, ranging from transcription to metabolism to genome stability. Thus, their dysregulation has been related to the pathogenesis of different diseases. In this review, we discussed the pharmacological approaches based on sirtuins’ modulators (both inhibitors and activators) that have been attempted in in vitro and/or in in vivo experimental settings, to highlight the therapeutic potential of targeting one/more specific sirtuin isoform(s) in cancer, neurodegenerative disorders and type 2 diabetes. Extensive research has already been performed to identify SIRT-1 and -2 modulators, while compounds targeting the other sirtuins have been less studied so far. Beside sections dedicated to each sirtuin, in the present review we also included sections dedicated to pan-sirtuins’ and to parasitic sirtuins’ modulators. A special focus is dedicated to the sirtuins’ modulators identified by the use of virtual screening.
Collapse
Affiliation(s)
- Elena Abbotto
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
- Correspondence:
| |
Collapse
|
8
|
Pan X, Pi C, Ruan X, Zheng H, Zhang D, Liu X. Mammalian Sirtuins and Their Relevance in Vascular Calcification. Front Pharmacol 2022; 13:907835. [PMID: 35677446 PMCID: PMC9168231 DOI: 10.3389/fphar.2022.907835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are a group of diseases with high morbidity and mortality that affect millions of people each year. Vascular calcification (VC) is an active process that involves the mineral deposition of calcium-phosphate complexes. VC is closely related to cardiovascular diseases, such as hypertension, heart failure, and calcific aortic stenosis, and is a type of ectopic calcification that occurs in the vessel walls. The sirtuins (silent mating-type information regulation 2; SIRTs), are a family of histone deacetylases whose function relies on nicotinamide adenine dinucleotide (NAD+). They have non-negligible functions in the regulation of energy metabolism, senescence, apoptosis, and other biological processes. Sirtuins have important effects on bone homeostasis and VC processes that share many similarities with bone formation. Sirtuins have been confirmed to deacetylate a variety of target proteins related to the occurrence and development of VC, thereby affecting the process of VC and providing new possibilities for the prevention and treatment of cardiovascular diseases. To facilitate the understanding of vascular calcification and accelerate the development of cardiovascular drugs, we reviewed and summarized recent research progress on the relationship between different types of sirtuins and VC.
Collapse
Affiliation(s)
- Xinyue Pan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xianchun Ruan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Hanhua Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| |
Collapse
|
9
|
Qu Y, Cao J, Wang D, Wang S, Li Y, Zhu Y. 14,15-Epoxyeicosatrienoic Acid Protect Against Glucose Deprivation and Reperfusion-Induced Cerebral Microvascular Endothelial Cells Injury by Modulating Mitochondrial Autophagy via SIRT1/FOXO3a Signaling Pathway and TSPO Protein. Front Cell Neurosci 2022; 16:888836. [PMID: 35558879 PMCID: PMC9086968 DOI: 10.3389/fncel.2022.888836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Neurovascular system plays a vital role in controlling the blood flow into brain parenchymal tissues. Additionally, it also facilitates the metabolism in neuronal biological activities. Cerebral microvascular endothelial cells (MECs) are involved in mediating progression of the diseases related to cerebral vessels, including stroke. Arachidonic acid can be transformed into epoxyeicosatrienoic acids (EETs) under the catalysis by cytochrome P450 epoxygenase. We have reported that EETs could protect neuronal function. In our research, the further role of 14,15-EET in the protective effects of cerebral MECs and the potential mechanisms involved in oxygen glucose deprivation and reperfusion (OGD/R) were elucidated. In our study, we intervened the SIRT1/FOXO3a pathway and established a TSPO knock down model by using RNA interference technique to explore the cytoprotective role of 14,15-EET in OGD/R injury. Cerebral MECs viability was remarkably reduced after OGD/R treatment, however, 14,15-EET could reverse this effect. To further confirm whether 14,15-EET was mediated by SIRT1/FOXO3a signaling pathway and translocator protein (TSPO) protein, we also detected autophagy-related proteins, mitochondrial membrane potential, apoptosis indicators, oxygen free radicals, etc. It was found that 14,15-EET could regulate the mitophagy induced by OGD/R. SIRT1/FOXO3a signaling pathway and TSPO regulation were related to the protective role of 14,15-EET in cerebral MECs. Moreover, we also explored the potential relationship between SIRT1/FOXO3a signaling pathway and TSPO protein. Our study revealed the protective role and the potential mechanisms of 14,15-EET in cerebral MECs under OGD/R condition.
Collapse
Affiliation(s)
- Youyang Qu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinlu Cao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shu Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yujie Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yulan Zhu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Durai P, Beeraka NM, Ramachandrappa HVP, Krishnan P, Gudur P, Raghavendra NM, Ravanappa PKB. Advances in PPARs Molecular Dynamics and Glitazones as a Repurposing Therapeutic Strategy through Mitochondrial Redox Dynamics against Neurodegeneration. Curr Neuropharmacol 2022; 20:893-915. [PMID: 34751120 PMCID: PMC9881103 DOI: 10.2174/1570159x19666211109141330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/20/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) activity has significant implications for the development of novel therapeutic modalities against neurodegenerative diseases. Although PPAR-α, PPAR-β/δ, and PPAR-γ nuclear receptor expressions are significantly reported in the brain, their implications in brain physiology and other neurodegenerative diseases still require extensive studies. PPAR signaling can modulate various cell signaling mechanisms involved in the cells contributing to on- and off-target actions selectively to promote therapeutic effects as well as the adverse effects of PPAR ligands. Both natural and synthetic ligands for the PPARα, PPARγ, and PPARβ/δ have been reported. PPARα (WY 14.643) and PPARγ agonists can confer neuroprotection by modulating mitochondrial dynamics through the redox system. The pharmacological effect of these agonists may deliver effective clinical responses by protecting vulnerable neurons from Aβ toxicity in Alzheimer's disease (AD) patients. Therefore, the current review delineated the ligands' interaction with 3D-PPARs to modulate neuroprotection, and also deciphered the efficacy of numerous drugs, viz. Aβ aggregation inhibitors, vaccines, and γ-secretase inhibitors against AD; this review elucidated the role of PPAR and their receptor isoforms in neural systems, and neurodegeneration in human beings. Further, we have substantially discussed the efficacy of PPREs as potent transcription factors in the brain, and the role of PPAR agonists in neurotransmission, PPAR gamma coactivator-1α (PGC-1α) and mitochondrial dynamics in neuroprotection during AD conditions. This review concludes with the statement that the development of novel PPARs agonists may benefit patients with neurodegeneration, mainly AD patients, which may help mitigate the pathophysiology of dementia, subsequently improving overall the patient's quality of life.
Collapse
Affiliation(s)
- Priya Durai
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | - Narasimha M. Beeraka
- Center of Excellence in Regenerative Medicine and Molecular Biology (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India;,I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119146, Russia
| | - Hemanth Vikram Poola Ramachandrappa
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | | | - Pranesh Gudur
- Swamy Vivekananda Yoga Anusandhana Samsthana Deemed University, Bengaluru 560 105, India
| | | | - Prashantha Kumar Bommenahally Ravanappa
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research, Mysuru, Karnataka, India;,Address correspondence to this author at the Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India; E-mail:
| |
Collapse
|
11
|
Huang C, Sharma A, Thakur R, Rai D, Katiki M, Germano JDF, Song Y, Singh S, Sin J, Sengstock D, Andres AM, Murali R, Mentzer RM, Gottlieb RA, Piplani H. Asporin, an extracellular matrix protein, is a beneficial regulator of cardiac remodeling. Matrix Biol 2022; 110:40-59. [DOI: 10.1016/j.matbio.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/29/2022] [Accepted: 04/19/2022] [Indexed: 01/15/2023]
|
12
|
Yang Y, Liu Y, Wang Y, Chao Y, Zhang J, Jia Y, Tie J, Hu D. Regulation of SIRT1 and Its Roles in Inflammation. Front Immunol 2022; 13:831168. [PMID: 35359990 PMCID: PMC8962665 DOI: 10.3389/fimmu.2022.831168] [Citation(s) in RCA: 244] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/15/2022] [Indexed: 12/28/2022] Open
Abstract
The silent information regulator sirtuin 1 (SIRT1) protein, a highly conserved NAD+-dependent deacetylase belonging to the sirtuin family, is a post-translational regulator that plays a role in modulating inflammation. SIRT1 affects multiple biological processes by deacetylating a variety of proteins including histones and non-histone proteins. Recent studies have revealed intimate links between SIRT1 and inflammation, while alterations to SIRT1 expression and activity have been linked to inflammatory diseases. In this review, we summarize the mechanisms that regulate SIRT1 expression, including upstream activators and suppressors that operate on the transcriptional and post-transcriptional levels. We also summarize factors that influence SIRT1 activity including the NAD+/NADH ratio, SIRT1 binding partners, and post-translational modifications. Furthermore, we underscore the role of SIRT1 in the development of inflammation by commenting on the proteins that are targeted for deacetylation by SIRT1. Finally, we highlight the potential for SIRT1-based therapeutics for inflammatory diseases.
Collapse
Affiliation(s)
- Yunshu Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yunwei Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yongyi Chao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jinxin Zhang
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jun Tie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
13
|
Ravaglia S, Malovini A, Cirio S, Danesino C, De Filippi P, Moggio M, Mongini T, Maggi L, Servidei S, Vianello A, Toscano A, Tonin P, Maioli MA, Parini R, Filosto M, Crescimanno G, Arceri S, Piran M, Carlucci A. Polymorphism in exercise genes and respiratory function in late-onset Pompe disease. J Appl Physiol (1985) 2021; 131:1762-1771. [PMID: 34734785 DOI: 10.1152/japplphysiol.00154.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genetic polymorphisms influencing muscle structure and metabolism may affect the phenotype of metabolic myopathies. We here analyze the possible influence of a wide panel of "exercise genes" on the severity and progression of respiratory dysfunction in late-onset Pompe disease (LOPD). We stratified patients with comparable age and disease duration according to the severity of their respiratory phenotype, assessed by both upright FVC% and postural drop in FVC%. We included 43 patients with LOPD (25 males, age 50.8 ± 13.6 yr) with a 2-yr follow-up since the beginning of enzyme replacement therapy (ERT). Twenty-two patients showed a postural drop >25% T0, seven other patients developed it during the follow-up. We analyzed the relationship between the progression of respiratory dysfunction and genetic polymorphisms affecting muscle function and structure [angiotensin converting enzyme (ACE), α-actinin 3 (ACTN3), peroxisome proliferator-activated receptor α (PPR-α), angiotensin (AGT)], glycogen metabolism [glycogen synthase (GYS), glycogen synthase kinase-3 isoform β (GSK3β)], and autophagy [sirtuin 1 (SIRT1), autophagy-related gene 7 (ATG7)]. Individuals carrying two copies of the ACE D-allele shared a 24-fold increase in the risk of severe respiratory dysfunction and progression during the 2-yr follow-up. ACTN3-XX polymorphism was also associated with worse respiratory outcome. The study of exercise genes is of particular interest in respiratory muscles, due to their peculiar features, that is, continuous, low-intensity contraction and prominent recruitment of type I fibers. In line with previous observations on skeletal muscles, ACE-DD and ACTN3-XX genotypes were associated with indirect evidence of more severe respiratory phenotypes. On the contrary, polymorphisms related to autophagy and glycogen metabolism did not seem to influence respiratory muscles.NEW & NOTEWORTHY Previous reports evaluated the role of exercise genes in influencing skeletal muscle phenotype and response to ERT in LOPD. Here, we investigate the role of polymorphisms in several exercise gene, focusing on respiratory muscles. ACE-DD and ACTN3-XX polymorphisms, possibly influencing muscle properties and fiber composition, were associated with more severe respiratory phenotypes.
Collapse
Affiliation(s)
| | - Alberto Malovini
- Riabilitazione respiratoria, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Serena Cirio
- Riabilitazione respiratoria, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Cesare Danesino
- Dipartimento di Medicina Molecolare, Unità di Biologia Generale e Genetica Medica, Università di Pavia, Pavia, Italy
| | - Paola De Filippi
- Dipartimento di Medicina Molecolare, Unità di Biologia Generale e Genetica Medica, Università di Pavia, Pavia, Italy
| | - Maurizio Moggio
- UO Malattie Neuromuscolari e Rare, IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Centro Dino Ferrari Univ Studi Milano, Milan, Italy
| | - Tiziana Mongini
- Neuromuscular Unit, Department of Neurosciences RLM, University of Torino, Torino, Italy
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serena Servidei
- Istituto di Neurologia, Università Cattolica Policlinico Gemelli, Rome, Italy
| | - Andrea Vianello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Antonio Toscano
- Department of Clinical and Experimental Medicine, UOC Neurologia e Malattie Neuromuscolari, University of Messina, Messina, Italy
| | - Paola Tonin
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, Verona, Italy
| | | | | | - Massimiliano Filosto
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,NeMO-Brescia Clinical Center for Neuromuscular Diseases, Brescia, Italy
| | - Grazia Crescimanno
- Regional Center for Prevention and Treatment of Respiratory Complications of Rare Genetic Neuromuscular Diseases, Villa Sofia-Cervello Hospital, Palermo, Italy
| | | | - Manuela Piran
- Riabilitazione respiratoria, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Annalisa Carlucci
- Riabilitazione respiratoria, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| |
Collapse
|
14
|
Blasl AT, Schulze S, Qin C, Graf LG, Vogt R, Lammers M. Post-translational lysine ac(et)ylation in health, ageing and disease. Biol Chem 2021; 403:151-194. [PMID: 34433238 DOI: 10.1515/hsz-2021-0139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
The acetylation/acylation (ac(et)ylation) of lysine side chains is a dynamic post-translational modification (PTM) regulating fundamental cellular processes with implications on the organisms' ageing process: metabolism, transcription, translation, cell proliferation, regulation of the cytoskeleton and DNA damage repair. First identified to occur on histones, later studies revealed the presence of lysine ac(et)ylation in organisms of all kingdoms of life, in proteins covering all essential cellular processes. A remarkable finding showed that the NAD+-dependent sirtuin deacetylase Sir2 has an impact on replicative lifespan in Saccharomyces cerevisiae suggesting that lysine acetylation has a direct role in the ageing process. Later studies identified sirtuins as mediators for beneficial effects of caloric/dietary restriction on the organisms' health- or lifespan. However, the molecular mechanisms underlying these effects are only incompletely understood. Progress in mass-spectrometry, structural biology, synthetic and semi-synthetic biology deepened our understanding of this PTM. This review summarizes recent developments in the research field. It shows how lysine ac(et)ylation regulates protein function, how it is regulated enzymatically and non-enzymatically, how a dysfunction in this post-translational machinery contributes to disease development. A focus is set on sirtuins and lysine acyltransferases as these are direct sensors and mediators of the cellular metabolic state. Finally, this review highlights technological advances to study lysine ac(et)ylation.
Collapse
Affiliation(s)
- Anna-Theresa Blasl
- Department Synthetic and Structural Biochemistry, Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487Greifswald, Germany
| | - Sabrina Schulze
- Department Synthetic and Structural Biochemistry, Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487Greifswald, Germany
| | - Chuan Qin
- Department Synthetic and Structural Biochemistry, Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487Greifswald, Germany
| | - Leonie G Graf
- Department Synthetic and Structural Biochemistry, Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487Greifswald, Germany
| | - Robert Vogt
- Department Synthetic and Structural Biochemistry, Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487Greifswald, Germany
| | - Michael Lammers
- Department Synthetic and Structural Biochemistry, Institute for Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487Greifswald, Germany
| |
Collapse
|
15
|
Biochemical insight into pseudouridine synthase 7 (PUS7) as a novel interactor of sirtuin, SIRT1. Biochem Biophys Res Commun 2019; 518:598-604. [PMID: 31451225 DOI: 10.1016/j.bbrc.2019.08.097] [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: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 01/28/2023]
Abstract
Sirtuin1 (SIRT1) forms a dynamic regulatory network with multiple proteins. The SIRT1 protein interactome comprises histone, non-histone substrates, and modulators of SIRT1 deacetylase. Proteomic studies have enlisted several proteins in SIRT1 network, but the structural and functional details of their interactions remain largely unexplored. In this study, we establish Pseudouridine synthase 7 (PUS7), a nuclear protein involved in stem cell development and intellectual disabilities, as a novel interactor of SIRT1. The binding regions are predicted and analyzed based on molecular docking studies. The direct interaction occurs between SIRT1 and PUS7, as evidenced by pull-down studies and surface plasmon resonance (SPR) assay. Furthermore, the truncation studies unambiguously suggested that the N-terminal region of PUS7 is essential for forming a stable complex with SIRT1. Overall, our results suggest that PUS7 may regulate the SIRT1 function when it directly interacts with SIRT1.
Collapse
|
16
|
Lermant A, Murdoch CE. Cysteine Glutathionylation Acts as a Redox Switch in Endothelial Cells. Antioxidants (Basel) 2019; 8:E315. [PMID: 31426416 PMCID: PMC6720164 DOI: 10.3390/antiox8080315] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Oxidative post-translational modifications (oxPTM) of receptors, enzymes, ion channels and transcription factors play an important role in cell signaling. oxPTMs are a key way in which oxidative stress can influence cell behavior during diverse pathological settings such as cardiovascular diseases (CVD), cancer, neurodegeneration and inflammatory response. In addition, changes in oxPTM are likely to be ways in which low level reactive oxygen and nitrogen species (RONS) may contribute to redox signaling, exerting changes in physiological responses including angiogenesis, cardiac remodeling and embryogenesis. Among oxPTM, S-glutathionylation of reactive cysteines emerges as an important regulator of vascular homeostasis by modulating endothelial cell (EC) responses to their local redox environment. This review summarizes the latest findings of S-glutathionylated proteins in major EC pathways, and the functional consequences on vascular pathophysiology. This review highlights the diversity of molecules affected by S-glutathionylation, and the complex consequences on EC function, thereby demonstrating an intricate dual role of RONS-induced S-glutathionylation in maintaining vascular homeostasis and participating in various pathological processes.
Collapse
Affiliation(s)
- Agathe Lermant
- Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland DD1 9SY, UK
| | - Colin E Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland DD1 9SY, UK.
| |
Collapse
|
17
|
Lin C, Liu M, Zhu X, Zhang M, Xu S, Wang D, Zhao Y. Cloning and expression of the lifespan-associated protein Sir2 from Daphnia pulex. Comp Biochem Physiol B Biochem Mol Biol 2019; 231:1-10. [DOI: 10.1016/j.cbpb.2019.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 11/15/2022]
|
18
|
Manna D, Bhuyan R, Ghosh R. Probing the mechanism of SIRT1 activation by a 1,4-dihydropyridine. J Mol Model 2018; 24:340. [PMID: 30448921 DOI: 10.1007/s00894-018-3877-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/05/2018] [Indexed: 11/25/2022]
Abstract
The NAD+-dependent deacetylase SIRT1 plays important roles in several physiological processes such as transcription, genome stability, stress responses, and aging. Due to its diverse role in metabolisms, SIRT1 has emerged as a potential therapeutic target in many human disorders such as type II diabetes, cardiovascular and neurodegenerative diseases, and cancer. Recent studies have reported that modulation of SIRT1 activity by phenolic activators like resveratrol and some 1,4-dihydropyridines (1,4-DHPs) can inhibit tumor growth by promoting apoptosis in cancer cells. However, the mechanism of SIRT1 activation is still not clear. In this report, we have tried to elucidate the mechanism of SIRT1 activation from studies on its interaction with a synthetic 1,4-DHP derivative (DHP-8; 3,5-diethoxy carbonyl-4-(4-nitrophenyl)-2,6-dimethyl-1,4-dihydropyridine) using molecular modeling, docking, simulation, and free energy analyses. Owing to the absence of full-length human SIRT1 structure, multi-template based modeling approach was opted followed by docking of DHP-8 at its allosteric site. In presence of DHP-8, the overall conformation of SIRT1 was found to be more stable (especially at its substrate binding sites) with a large structural variation at its N-terminal domain while bound to substrate p53 or p53-W. Determination of the MM/PBSA free energy indicated that the binding of DHP-8 to SIRT1 significantly increased the binding affinity of SIRT1 to its substrate p53-W as well as to NAD+. Overall, this study depicts the atomistic detailed mechanism for the direct activation of SIRT1 by a 1,4-DHP. This would serve to develop new SIRT1 activators for future therapeutic perspectives.
Collapse
Affiliation(s)
- Debashri Manna
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Rajabrata Bhuyan
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Rita Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India.
| |
Collapse
|
19
|
Fujita Y, Yamashita T. Sirtuins in Neuroendocrine Regulation and Neurological Diseases. Front Neurosci 2018; 12:778. [PMID: 30416425 PMCID: PMC6213750 DOI: 10.3389/fnins.2018.00778] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Silent information regulator 1 (SIRT1) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Sirtuin was originally studied as the lifespan-extending gene, silent information regulator 2 (SIRT2) in budding yeast. There are seven mammalian homologs of sirtuin (SIRT1–7), and SIRT1 is the closest homolog to SIRT2. SIRT1 modulates various key targets via deacetylation. In addition to histones, these targets include transcription factors, such as forkhead box O (FOXO), Ku70, p53, NF-κB, PPAR-gamma co-activator 1-alpha (PGC-1α), and peroxisome proliferator-activated receptor γ (PPARγ). SIRT1 has many biological functions, including aging, cell survival, differentiation, and metabolism. Genetic and physiological analyses in animal models have shown beneficial roles for SIRT1 in the brain during both development and adulthood. Evidence from in vivo and in vitro studies have revealed that SIRT1 regulates the cellular fate of neural progenitors, axon elongation, dendritic branching, synaptic plasticity, and endocrine function. In addition to its importance in physiological processes, SIRT1 has also been implicated in protection of neurons from degeneration in models of neurological diseases, such as traumatic brain injury and Alzheimer’s disease. In this review, we focus on the role of SIRT1 in the neuroendocrine system and neurodegenerative diseases. We also discuss the potential therapeutic implications of targeting the sirtuin pathway.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
20
|
Ong AL, Ramasamy TS. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res Rev 2018; 43:64-80. [PMID: 29476819 DOI: 10.1016/j.arr.2018.02.004] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/23/2018] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
Regulatory role of Sirtuin 1 (SIRT1), one of the most extensively studied members of its kind in histone deacetylase family in governing multiple cellular fates, is predominantly linked to p53 activity. SIRT1 deacetylates p53 in a NAD+-dependent manner to inhibit transcription activity of p53, in turn modulate pathways that are implicated in regulation of tissue homoeostasis and many disease states. In this review, we discuss the role of SIRT1-p53 pathway and its regulatory axis in the cellular events which are implicated in cellular aging, cancer and reprogramming. It is noteworthy that these cellular events share few common regulatory pathways, including SIRT1-p53-LDHA-Myc, miR-34a,-Let7 regulatory network, which forms a positive feedback loop that controls cell cycle, metabolism, proliferation, differentiation, epigenetics and many others. In the context of aging, SIRT1 expression is reduced as a protective mechanism against oncogenesis and for maintenance of tissue homeostasis. Interestingly, its activation in aged cells is evidenced in response to DNA damage to protect the cells from p53-dependent apoptosis or senescence, predispose these cells to neoplastic transformation. Importantly, the dual roles of SIRT1-p53 axis in aging and tumourigenesis, either as tumour suppressor or tumour promoter are determined by SIRT1 localisation and type of cells. Conceptualising the distinct similarity between tumorigenesis and cellular reprogramming, this review provides a perspective discussion on involvement of SIRT1 in improving efficiency in the induction and maintenance of pluripotent state. Further research in understanding the role of SIRT1-p53 pathway and their associated regulators and strategies to manipulate this regulatory axis very likely foster the development of therapeutics and strategies for treating cancer and aging-associated degenerative diseases.
Collapse
|
21
|
Steinmetz C, Kashyap A, Zhivkova N, Alizor H, Ernst I, Gottfried-Brand D, Janssen H, Teufel A, Schulze-Bergkamen H, Lotz J, Kuball J, Theobald M, Heise M, Lang H, Galle PR, Strand D, Strand S. Activation of silent mating type information regulation 2 homolog 1 by human chorionic gonadotropin exerts a therapeutic effect on hepatic injury and inflammation. Hepatology 2017; 65:2074-2089. [PMID: 28108987 DOI: 10.1002/hep.29072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/30/2022]
Abstract
UNLABELLED Incidence and prevalence of inflammatory liver diseases has increased over the last years, but therapeutic options are limited. Pregnancy induces a state of immune tolerance, which can result in spontaneous improvement of clinical symptoms of certain autoimmune diseases including autoimmune hepatitis (AIH). We investigated the immune-suppressive mechanisms of the human pregnancy hormone, chorionic gonadotropin (hCG), in the liver. hCG signaling activates silent mating type information regulation 2 homolog 1 (SIRT1), which deacetylates forkhead box o3 (FOXO3a), leading to repression of proapoptotic gene expression, because the immunosuppressive consequence attributed to the absence of caspase-3 activity of hepatocellular interleukin 16 (IL-16) is no longer processed and released. Thus, serum levels of IL-16, a key chemotactic factor for CD4+ lymphocytes, were reduced and migration to injured hepatocytes prevented. Furthermore, elevated IL-16 levels are found in the sera from patients with AIH, hepatitis B virus, hepatitis C virus, and nonalcoholic steatohepatitis. CONCLUSION Here, we report that hCG regulates the SIRT1/FOXO3a axis in hepatocytes, resulting in immune suppression by attenuating caspase-3-dependent IL-16 processing and release, which concomitantly prevents autoaggressive T-cell infiltration of the liver. Considering the low toxicity profile of hCG in humans, interrupting the inflammatory cycle by hCG opens new perspectives for therapeutic intervention of inflammatory liver diseases. (Hepatology 2017;65:2074-2089).
Collapse
Affiliation(s)
- Caroline Steinmetz
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Anubha Kashyap
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Nataliya Zhivkova
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Henry Alizor
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Isabell Ernst
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | | | - Henning Janssen
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Teufel
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | | | - Johannes Lotz
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Jürgen Kuball
- Department of Immunology, Department of Hematology and Van Creveld Clinic University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthias Theobald
- Department of Hematology and Oncology, Johannes Gutenberg University, Mainz, Germany
| | - Michael Heise
- General, Visceral and Transplant Surgery, Johannes Gutenberg-University, Mainz, Germany
| | - Hauke Lang
- General, Visceral and Transplant Surgery, Johannes Gutenberg-University, Mainz, Germany
| | - Peter R Galle
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Dennis Strand
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Susanne Strand
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
22
|
Mahmoud AA, Moghazy HM, Ezat MA. Association of sirtuin 1 gene single nucleotide polymorphisms with type 2 diabetes mellitus in essential hypertension patients. Meta Gene 2016. [DOI: 10.1016/j.mgene.2016.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
23
|
Chen GD, Yu WD, Chen XP. SirT1 activator represses the transcription of TNF‑α in THP‑1 cells of a sepsis model via deacetylation of H4K16. Mol Med Rep 2016; 14:5544-5550. [PMID: 27878240 PMCID: PMC5355689 DOI: 10.3892/mmr.2016.5942] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 10/12/2016] [Indexed: 01/27/2023] Open
Abstract
Sepsis is a systemic inflammatory response resulting from the excessive production of pro-inflammatory cytokines, including tumor necrosis factor (TNF)‑α. Sirtuin 1 (SirT1) actively deacetylates histone proteins, and facilitates chromatin compaction and gene silencing. In the present study, a cell model of sepsis, comprising lipopolysaccharide (LPS)‑tolerant THP‑1 cells, was used to investigate whether the SirT1 activator, resveratrol, repressed the transcription of TNF‑α. Chromatin immunoprecipitation and real‑time PCR were used to determine the transcription of the TNF‑α promoter. The result revealed that the binding of SirT1 to the TNF‑α promoter was decreased by LPS stimulation in normal cells. However, in LPS‑tolerant cells, nuclear protein levels of SirT1 remained elevated, and LPS stimulation had no significant effect on the binding of SirT1 to the TNF‑α promoter. However, the activity of SirT1 was increased and binding of ace‑H4K16 to the TNF‑α promoter was decreased with resveratrol treatment in the tolerant cells. It was concluded that resveratrol stimulated sirtuin activity in LPS‑tolerant THP‑1 cells, and repressed TNF‑α transcription through the deacetylation of H4K16, without affecting the methylation of H3K9. Resveratrol offers potential as an infective candidate to alleviate inflammation in patients with sepsis.
Collapse
Affiliation(s)
- Guo-Dong Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Wei-Dong Yu
- Central Laboratory, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Xiao-Ping Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Nosocomial Infection, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| |
Collapse
|
24
|
Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun 2016; 483:1166-1177. [PMID: 27514452 DOI: 10.1016/j.bbrc.2016.08.043] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/21/2016] [Accepted: 08/07/2016] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors and they serve to be a promising therapeutic target for several neurodegenerative disorders, which includes Parkinson disease, Alzheimer's disease, Huntington disease and Amyotrophic Lateral Sclerosis. PPARs play an important role in the downregulation of mitochondrial dysfunction, proteasomal dysfunction, oxidative stress, and neuroinflammation, which are the major causes of the pathogenesis of neurodegenerative disorders. In this review, we discuss about the role of PPARs as therapeutic targets in neurodegenerative disorders. Several experimental approaches suggest potential application of PPAR agonist as well as antagonist in the treatment of neurodegenerative disorders. Several epidemiological studies found that the regular usage of PPAR activating non-steroidal anti-inflammatory drugs is effective in decreasing the progression of neurodegenerative diseases including PD and AD. We also reviewed the neuroprotective effects of PPAR agonists and associated mechanism of action in several neurodegenerative disorders both in vitro as well as in vivo animal models.
Collapse
Affiliation(s)
- Swati Agarwal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India
| | - Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India.
| |
Collapse
|
25
|
Subramaniyan B, Jagadeesan K, Ramakrishnan S, Mathan G. Targeting the interaction of Aurora kinases and SIRT1 mediated by Wnt signaling pathway in colorectal cancer: A critical review. Biomed Pharmacother 2016; 82:413-24. [DOI: 10.1016/j.biopha.2016.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/18/2016] [Accepted: 05/18/2016] [Indexed: 12/22/2022] Open
|
26
|
Kumar A, Chauhan S. How much successful are the medicinal chemists in modulation of SIRT1: A critical review. Eur J Med Chem 2016; 119:45-69. [PMID: 27153347 DOI: 10.1016/j.ejmech.2016.04.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/14/2016] [Accepted: 04/25/2016] [Indexed: 12/27/2022]
Abstract
Silent information regulator two homologue one (SIRT1) is the most widely studied member of the sirtuin family related to histone deacetylases class III super-family using nicotinamide adenine dinucleotide (NAD(+)) as its cofactor. It is located in the nucleus but also modulates the targets in cytoplasm and mainly acts as transacetylase rather than deacetylase. SIRT1 specifically cleaves the nicotinamide ribosyl bond of NAD(+) and transfers the acetyl group from proteins to their co-substrate through an ADP- ribose-peptidyl imidate intermediate. It has been indicated that SIRT1 and its histone as well as non histone targets are involved in a wide range of biological courses including metabolic diseases, age related diseases, viral infection, inflammation, tumor-cell growth and metastasis. Modulation of SIRT1 expression may present a new insight in the discovery of a number of therapeutics. This review summarizes studies about SIRT1 and mainly focuses on the various modulators of SIRT1 evolved by natural as well as synthetic means.
Collapse
Affiliation(s)
- Ashwani Kumar
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India.
| | - Shilpi Chauhan
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| |
Collapse
|
27
|
Sirtuin 1 gene rs2273773 C>T single nucleotide polymorphism and protein oxidation markers in asthmatic patients. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2016. [DOI: 10.1016/j.ejmhg.2015.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
28
|
Potential Modulation of Sirtuins by Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:9831825. [PMID: 26788256 PMCID: PMC4691645 DOI: 10.1155/2016/9831825] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/20/2015] [Indexed: 12/13/2022]
Abstract
Sirtuins are a conserved family of NAD-dependent protein deacylases. Initially proposed as histone deacetylases, it is now known that they act on a variety of proteins including transcription factors and metabolic enzymes, having a key role in the regulation of cellular homeostasis. Seven isoforms are identified in mammals (SIRT1–7), all of them sharing a conserved catalytic core and showing differential subcellular localization and activities. Oxidative stress can affect the activity of sirtuins at different levels: expression, posttranslational modifications, protein-protein interactions, and NAD levels. Mild oxidative stress induces the expression of sirtuins as a compensatory mechanism, while harsh or prolonged oxidant conditions result in dysfunctional modified sirtuins more prone to degradation by the proteasome. Oxidative posttranslational modifications have been identified in vitro and in vivo, in particular cysteine oxidation and tyrosine nitration. In addition, oxidative stress can alter the interaction with other proteins, like SIRT1 with its protein inhibitor DBC1 resulting in a net increase of deacetylase activity. In the same way, manipulation of cellular NAD levels by pharmacological inhibition of other NAD-consuming enzymes results in activation of SIRT1 and protection against obesity-related pathologies. Nevertheless, further research is needed to establish the molecular mechanisms of redox regulation of sirtuins to further design adequate pharmacological interventions.
Collapse
|
29
|
Bergantino F, Guariniello S, Raucci R, Colonna G, De Luca A, Normanno N, Costantini S. Structure–fluctuation–function relationships of seven pro-angiogenic isoforms of VEGFA, important mediators of tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:410-25. [DOI: 10.1016/j.bbapap.2015.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/06/2015] [Accepted: 01/14/2015] [Indexed: 10/24/2022]
|
30
|
Vyas VK, Goel A, Ghate M, Patel P. Ligand and structure-based approaches for the identification of SIRT1 activators. Chem Biol Interact 2015; 228:9-17. [PMID: 25595223 DOI: 10.1016/j.cbi.2015.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/05/2014] [Accepted: 01/02/2015] [Indexed: 01/18/2023]
Abstract
SIRT1 is a NAD(+)-dependent deacetylase that involved in various important metabolic pathways. Combined ligand and structure-based approach was utilized for identification of SIRT1 activators. Pharmacophore models were developed using DISCOtech and refined with GASP module of Sybyl X software. Pharmacophore models were composed of two hydrogen bond acceptor (HBA) atoms, two hydrogen bond donor (HBD) sites and one hydrophobic (HY) feature. The pharmacophore models were validated through receiver operating characteristic (ROC) and Güner-Henry (GH) scoring methods. Model-2 was selected as best model among the model 1-3, based on ROC and GH score value, and found reliable in identification of SIRT1 activators. Model-2 (3D search query) was searched against Zinc database. Several compounds with different chemical scaffold were retrieved as hits. Currently, there is no experimental SIRT1 3D structure available, therefore, we modeled SIRT1 protein structure using homology modeling. Compounds with Qfit value of more than 86 were selected for docking study into the SIRT1 homology model to explore the binding mode of retrieved hits in the active allosteric site. Finally, in silico ADMET prediction study was performed with two best docked compounds. Combination of ligand and structure-based modeling methods identified active hits, which may be good lead compounds to develop novel SIRT1 activators.
Collapse
Affiliation(s)
- Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India.
| | - Ashutosh Goel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India
| | - Manjunath Ghate
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India
| | - Palak Patel
- Institute of Science, Nirma University, Ahmedabad 382 481, Gujarat, India
| |
Collapse
|
31
|
Pulla VK, Alvala M, Sriram DS, Viswanadha S, Sriram D, Yogeeswari P. Structure-based drug design of small molecule SIRT1 modulators to treat cancer and metabolic disorders. J Mol Graph Model 2014; 52:46-56. [DOI: 10.1016/j.jmgm.2014.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 05/05/2014] [Accepted: 06/17/2014] [Indexed: 11/29/2022]
|
32
|
Guariniello S, Colonna G, Raucci R, Costantini M, Di Bernardo G, Bergantino F, Castello G, Costantini S. Structure-function relationship and evolutionary history of the human selenoprotein M (SelM) found over-expressed in hepatocellular carcinoma. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1844:447-456. [PMID: 24332979 DOI: 10.1016/j.bbapap.2013.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/28/2013] [Accepted: 12/02/2013] [Indexed: 11/17/2022]
Abstract
In humans we know 25 selenoproteins that play important roles in redox regulation, detoxification, immune-system protection and viral suppression. In particular, selenoprotein M (SelM) may function as thiol disulfide oxidoreductase that participates in the formation of disulfide bonds, and can be implicated in calcium responses. However, it presents a redox motif (CXXU), where U is a selenocysteine, and may also function as redox regulator because its decreased or increased expression regulated by dietary selenium alters redox homeostasis. No data are reported in literature about its involvement in cancer but only in neurodegenerative diseases. In this paper we evaluated the SelM expression in two hepatoma cell lines, HepG2 and Huh7, compared to normal hepatocytes. The results suggested its involvement in hepatocellular carcinoma (HCC) as well as its possible use to follow the progression of this cancer as putative marker. The aim of this study has been to analyze the structure-function relationships of SelM. Hence, firstly we studied the evolutionary history of this protein by phylogenetic analysis and GC content of genes from various species. So, we modeled the three-dimensional structure of the human SelM evaluating its energetic stability by molecular dynamics simulations. Moreover, we modeled some of its mutants to obtain structural information helpful for structure-based drug design.
Collapse
Affiliation(s)
- Stefano Guariniello
- Biochemistry, Biophysics and General Pathology Department and Computational Biology Doctorate, Second University of Naples, Naples, Italy
| | - Giovanni Colonna
- Biochemistry, Biophysics and General Pathology Department and Computational Biology Doctorate, Second University of Naples, Naples, Italy
| | - Raffaele Raucci
- Biochemistry, Biophysics and General Pathology Department and Computational Biology Doctorate, Second University of Naples, Naples, Italy
| | | | - Gianni Di Bernardo
- Department of Experimental Medicine, Section of Biotechnology and Molecular Biology, Faculty of Medicine, Second University of Naples, Naples, Italy
| | - Francesca Bergantino
- Pharmacogenomic Laboratory, Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Per Lo Studio E La Cura Dei Tumori "Fondazione Giovanni Pascale", IRCCS, Italy
| | - Giuseppe Castello
- Istituto Nazionale Per Lo Studio E La Cura Dei Tumori "Fondazione Giovanni Pascale", IRCCS, Italy
| | - Susan Costantini
- Istituto Nazionale Per Lo Studio E La Cura Dei Tumori "Fondazione Giovanni Pascale", IRCCS, Italy.
| |
Collapse
|
33
|
Costantini S, Raucci R, Colonna G, Mercurio FA, Trotta AM, Paola R, Leone M, Rossi F, Pellegrino C, Castello G, Scala S. Peptides targeting chemokine receptor CXCR4: structural behavior and biological binding studies. J Pept Sci 2014; 20:270-8. [DOI: 10.1002/psc.2614] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/20/2013] [Accepted: 12/20/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Susan Costantini
- Istituto Nazionale per lo Studio e la Cura dei Tumori ‘Fondazione Giovanni Pascale’ (IRCCS); Naples Italy
| | - Raffaele Raucci
- Department of Biochemistry, Biophysics and General Pathology; Second University of Naples; Naples Italy
| | - Giovanni Colonna
- Department of Biochemistry, Biophysics and General Pathology; Second University of Naples; Naples Italy
| | - Flavia Anna Mercurio
- Department of Pharmacy; University of Naples ‘Federico II’; Naples Italy
- CIRPeB; Via Mezzocannone 16 Naples Italy
- Institute of Biostructures and Bioimaging, CNR; Naples Italy
| | - Anna Maria Trotta
- Istituto Nazionale per lo Studio e la Cura dei Tumori ‘Fondazione Giovanni Pascale’ (IRCCS); Naples Italy
| | - Ringhieri Paola
- Department of Pharmacy; University of Naples ‘Federico II’; Naples Italy
- CIRPeB; Via Mezzocannone 16 Naples Italy
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging, CNR; Naples Italy
| | - Filomena Rossi
- Department of Pharmacy; University of Naples ‘Federico II’; Naples Italy
- CIRPeB; Via Mezzocannone 16 Naples Italy
- Institute of Biostructures and Bioimaging, CNR; Naples Italy
| | - Carmela Pellegrino
- Istituto Nazionale per lo Studio e la Cura dei Tumori ‘Fondazione Giovanni Pascale’ (IRCCS); Naples Italy
| | - Giuseppe Castello
- Istituto Nazionale per lo Studio e la Cura dei Tumori ‘Fondazione Giovanni Pascale’ (IRCCS); Naples Italy
| | - Stefania Scala
- Istituto Nazionale per lo Studio e la Cura dei Tumori ‘Fondazione Giovanni Pascale’ (IRCCS); Naples Italy
| |
Collapse
|
34
|
Knight JRP, Allison SJ, Milner J. Active regulator of SIRT1 is required for cancer cell survival but not for SIRT1 activity. Open Biol 2013; 3:130130. [PMID: 24258275 PMCID: PMC3843821 DOI: 10.1098/rsob.130130] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/25/2013] [Indexed: 01/30/2023] Open
Abstract
The NAD(+)-dependent deacetylase SIRT1 is involved in diverse cellular processes, and has also been linked with multiple disease states. Among these, SIRT1 expression negatively correlates with cancer survival in both laboratory and clinical studies. Active regulator of SIRT1 (AROS) was the first reported post-transcriptional regulator of SIRT1 activity, enhancing SIRT1-mediated deacetylation and downregulation of the SIRT1 target p53. However, little is known regarding the role of AROS in regulation of SIRT1 during disease. Here, we report the cellular and molecular effects of RNAi-mediated AROS suppression, comparing this with the role of SIRT1 in a panel of human cell lines of both cancerous and non-cancerous origins. Unexpectedly, AROS is found to vary in its modulation of p53 acetylation according to cell context. AROS suppresses p53 acetylation only following the application of cell damaging stress, whereas SIRT1 suppresses p53 under all conditions analysed. This supplements the original characterization of AROS but indicates that SIRT1 activity can persist following suppression of AROS. We also demonstrate that knockdown of AROS induces apoptosis in three cancer cell lines, independent of p53 activation. Importantly, AROS is not required for the viability of three non-cancer cell lines indicating a putative role for AROS in specifically promoting cancer cell survival.
Collapse
|
35
|
Abstract
NAD(+)-dependent deacetylase SIRT1 is a master regulator of nucleosome positioning and chromatin structure, thereby reprogramming gene expression. In acute inflammation, chromatin departs from, and returns to, homeostasis in an orderly sequence. This sequence depends on shifts in NAD(+) availability for SIRT1 activation and deacetylation of signaling proteins, which support orderly gene reprogramming during acute inflammation by switching between euchromatin and heterochromatin. In contrast, in chronic inflammation and cancer, limited availability of NAD(+) and reduced expression of SIRT1 may sustain aberrant chromatin structure and functions. SIRT1 also influences inflammation and cancer by directly deacetylating targets like NFκB p65 and p53. Here, we review SIRT1 in the context of inflammation and cancer.
Collapse
Affiliation(s)
- Tie Fu Liu
- Molecular Medicine Section, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
36
|
McBurney MW, Clark-Knowles KV, Caron AZ, Gray DA. SIRT1 is a Highly Networked Protein That Mediates the Adaptation to Chronic Physiological Stress. Genes Cancer 2013; 4:125-34. [PMID: 24020004 DOI: 10.1177/1947601912474893] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
SIRT1 is a NAD(+)-dependent protein deacetylase that has a very large number of established protein substrates and an equally impressive list of biological functions thought to be regulated by its activity. Perhaps as notable is the remarkable number of points of conflict concerning the role of SIRT1 in biological processes. For example, evidence exists suggesting that SIRT1 is a tumor suppressor, is an oncogene, or has no effect on oncogenesis. Similarly, SIRT1 is variably reported to induce, inhibit, or have no effect on autophagy. We believe that the resolution of many conflicting results is possible by considering recent reports indicating that SIRT1 is an important hub interacting with a complex network of proteins that collectively regulate a wide variety of biological processes including cancer and autophagy. A number of the interacting proteins are themselves hubs that, like SIRT1, utilize intrinsically disordered regions for their promiscuous interactions. Many studies investigating SIRT1 function have been carried out on cell lines carrying undetermined numbers of alterations to the proteins comprising the SIRT1 network or on inbred mouse strains carrying fixed mutations affecting some of these proteins. Thus, the effects of modulating SIRT1 amount and/or activity are importantly determined by the genetic background of the cell (or the inbred strain of mice), and the effects attributed to SIRT1 are synthetic with the background of mutations and epigenetic differences between cells and organisms. Work on mice carrying alterations to the Sirt1 gene suggests that the network in which SIRT1 functions plays an important role in mediating physiological adaptation to various sources of chronic stress such as calorie restriction and calorie overload. Whether the catalytic activity of SIRT1 and the nuclear concentration of the co-factor, NAD(+), are responsible for modulating this activity remains to be determined. However, the effect of modulating SIRT1 activity must be interpreted in the context of the cell or tissue under investigation. Indeed, for SIRT1, we argue that context is everything.
Collapse
Affiliation(s)
- Michael W McBurney
- Program in Cancer Therapeutics, Ottawa Hospital Research Institute ; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | | | | |
Collapse
|
37
|
Sharma A, Costantini S, Colonna G. The protein-protein interaction network of the human Sirtuin family. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:1998-2009. [PMID: 23811471 DOI: 10.1016/j.bbapap.2013.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 05/31/2013] [Accepted: 06/18/2013] [Indexed: 12/15/2022]
Abstract
Protein-protein interaction networks are useful for studying human diseases and to look for possible health care through a holistic approach. Networks are playing an increasing and important role in the understanding of physiological processes such as homeostasis, signaling, spatial and temporal organizations, and pathological conditions. In this article we show the complex system of interactions determined by human Sirtuins (Sirt) largely involved in many metabolic processes as well as in different diseases. The Sirtuin family consists of seven homologous Sirt-s having structurally similar cores but different terminal segments, being rather variable in length and/or intrinsically disordered. Many studies have determined their cellular location as well as biological functions although molecular mechanisms through which they act are actually little known therefore, the aim of this work was to define, explore and understand the Sirtuin-related human interactome. As a first step, we have integrated the experimentally determined protein-protein interactions of the Sirtuin-family as well as their first and second neighbors to a Sirtuin-related sub-interactome. Our data showed that the second-neighbor network of Sirtuins encompasses 25% of the entire human interactome, and exhibits a scale-free degree distribution and interconnectedness among top degree nodes. Moreover, the Sirtuin sub interactome showed a modular structure around the core comprising mixed functions. Finally, we extracted from the Sirtuin sub-interactome subnets related to cancer, aging and post-translational modifications for information on key nodes and topological space of the subnets in the Sirt family network.
Collapse
Affiliation(s)
- Ankush Sharma
- Biochemistry, Biophysics and General Pathology Department, Second University of Naples, Naples, Italy
| | | | | |
Collapse
|
38
|
Costantini S, Raucci R, De Vero T, Castello G, Colonna G. Common structural interactions between the receptors CXCR3, CXCR4 and CXCR7 complexed with their natural ligands, CXCL11 and CXCL12, by a modeling approach. Cytokine 2013; 64:316-21. [PMID: 23773308 DOI: 10.1016/j.cyto.2013.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 04/07/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
Abstract
Chemokine receptor trio composed by CXCR3, CXCR4 and CXCR7 represents a hard and interesting challenge for cancer biology because these three receptors are found to be over-expressed in different cancers as well as to bind the same chemokines. In fact, CXCR4 interacts with CXCL12, CXCR7 not only with CXCL12 but also with CXCL11, that is a natural ligand for CXCR3. For these reasons, it seems necessary to define and to identify the structural determinants of CXCR3, CXCR4 and CXCR7 and their related physic-chemical properties that permit them to bind CXCL11 and CXCL12. Hence in this paper we show the modeling of CXCR7 and its complex with CXCL11 and CXCL12 compared to CXCR3/CXCL11 and CXCR4/CXCL12. Our results show that (i) CXCR3, CXCR4 and CXCR7 present similar trans-membrane helices and different conformations of N-terminal and C-terminal regions as well as of three extracellular loops, and (ii) the predominant interaction between the three receptors and the two chemokines are on hydrophobic and electrostatic basis. Moreover, our data confirm that CXCL12 binds to CXCR7 with higher affinity than to CXCR4. Methodologically, we can also conclude that our computational strategy is adequate to model correctly the interactions between these chemokines and their receptors; therefore, our models represent a good structural basis to design and develop peptides able to block contemporaneously CXCR3, CXCR4 and CXCR7 receptor trio.
Collapse
Affiliation(s)
- Susan Costantini
- Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione Giovanni Pascale, IRCCS, Italy.
| | | | | | | | | |
Collapse
|
39
|
Molecular architecture of the human protein deacetylase Sirt1 and its regulation by AROS and resveratrol. Biosci Rep 2013; 33:BSR20120121. [PMID: 23548308 PMCID: PMC3654483 DOI: 10.1042/bsr20120121] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sirtuins are NAD+-dependent protein deacetylases regulating metabolism, stress responses and ageing processes. Among the seven mammalian Sirtuins, Sirt1 is the physiologically best-studied isoform. It regulates nuclear functions such as chromatin remodelling and gene transcription, and it appears to mediate beneficial effects of a low calorie diet which can partly be mimicked by the Sirt1 activating polyphenol resveratrol. The molecular details of Sirt1 domain architecture and regulation, however, are little understood. It has a unique N-terminal domain and CTD (C-terminal domain) flanking a conserved Sirtuin catalytic core and these extensions are assumed to mediate Sirt1-specific features such as homo-oligomerization and activation by resveratrol. To analyse the architecture of human Sirt1 and functions of its N- and C-terminal extensions, we recombinantly produced Sirt1 and Sirt1 deletion constructs as well as the AROS (active regulator of Sirt1) protein. We then studied Sirt1 features such as molecular size, secondary structure and stimulation by small molecules and AROS. We find that Sirt1 is monomeric and has extended conformations in its flanking domains, likely disordered especially in the N-terminus, resulting in an increased hydrodynamic radius. Nevertheless, both termini increase Sirt1 deacetylase activity, indicating a regulatory function. We also find an unusual but defined conformation for AROS protein, which fails, however, to stimulate Sirt1. Resveratrol, in contrast, activates the Sirt1 catalytic core independent of the terminal domains, indicating a binding site within the catalytic core and suggesting that small molecule activators for other isoforms might also exist.
Collapse
|
40
|
Biason-Lauber A, Böni-Schnetzler M, Hubbard BP, Bouzakri K, Brunner A, Cavelti-Weder C, Keller C, Meyer-Böni M, Meier DT, Brorsson C, Timper K, Leibowitz G, Patrignani A, Bruggmann R, Boily G, Zulewski H, Geier A, Cermak JM, Elliott P, Ellis JL, Westphal C, Knobel U, Eloranta JJ, Kerr-Conte J, Pattou F, Konrad D, Matter CM, Fontana A, Rogler G, Schlapbach R, Regairaz C, Carballido JM, Glaser B, McBurney MW, Pociot F, Sinclair DA, Donath MY. Identification of a SIRT1 mutation in a family with type 1 diabetes. Cell Metab 2013; 17:448-455. [PMID: 23473037 PMCID: PMC3746172 DOI: 10.1016/j.cmet.2013.02.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/30/2012] [Accepted: 02/07/2013] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes is caused by autoimmune-mediated β cell destruction leading to insulin deficiency. The histone deacetylase SIRT1 plays an essential role in modulating several age-related diseases. Here we describe a family carrying a mutation in the SIRT1 gene, in which all five affected members developed an autoimmune disorder: four developed type 1 diabetes, and one developed ulcerative colitis. Initially, a 26-year-old man was diagnosed with the typical features of type 1 diabetes, including lean body mass, autoantibodies, T cell reactivity to β cell antigens, and a rapid dependence on insulin. Direct and exome sequencing identified the presence of a T-to-C exchange in exon 1 of SIRT1, corresponding to a leucine-to-proline mutation at residue 107. Expression of SIRT1-L107P in insulin-producing cells resulted in overproduction of nitric oxide, cytokines, and chemokines. These observations identify a role for SIRT1 in human autoimmunity and unveil a monogenic form of type 1 diabetes.
Collapse
Affiliation(s)
- Anna Biason-Lauber
- Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Marianne Böni-Schnetzler
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Basil P. Hubbard
- Department of Genetics, Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - Karim Bouzakri
- Department of Genetic Medicine and Development, University of Geneva, University Medical Center, 1211 Geneva, Switzerland
| | - Andrea Brunner
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Claudia Cavelti-Weder
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Cornelia Keller
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Monika Meyer-Böni
- Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Daniel T. Meier
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Caroline Brorsson
- Glostrup Research Institute, Glostrup University Hospital, 2600 Glostrup, Denmark
| | - Katharina Timper
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Gil Leibowitz
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Andrea Patrignani
- Functional Genomic Center Zurich, Federal Institute of Technology University of Zurich, 8057 Zurich, Switzerland
| | - Remy Bruggmann
- Functional Genomic Center Zurich, Federal Institute of Technology University of Zurich, 8057 Zurich, Switzerland
| | - Gino Boily
- University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Henryk Zulewski
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Andreas Geier
- Gastroenterology and Hepatology Center of Integrative Human Physiology and University Hospital Zurich, 8091 Zurich, Switzerland
| | | | | | | | | | - Urs Knobel
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Jyrki J. Eloranta
- Department of Clinical Pharmacology and Toxicology Center of Integrative Human Physiology and University Hospital Zurich, 8091 Zurich, Switzerland
| | | | | | - Daniel Konrad
- University Children's Hospital, 8032 Zurich, Switzerland
| | - Christian M. Matter
- Department of Cardiology Center of Integrative Human Physiology and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Adriano Fontana
- Institute of Experimental Immunology University of Zurich, 8057 Zurich, Switzerland
| | - Gerhard Rogler
- Gastroenterology and Hepatology Center of Integrative Human Physiology and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Ralph Schlapbach
- Functional Genomic Center Zurich, Federal Institute of Technology University of Zurich, 8057 Zurich, Switzerland
| | - Camille Regairaz
- Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | | | - Benjamin Glaser
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah Hebrew University Medical Center, 91120 Jerusalem, Israel
| | | | - Flemming Pociot
- Glostrup Research Institute, Glostrup University Hospital, 2600 Glostrup, Denmark
| | - David A. Sinclair
- Department of Genetics, Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - Marc Y. Donath
- Endocrinology, Diabetes, and Metabolism, and Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
41
|
Costantini S, Sharma A, Raucci R, Costantini M, Autiero I, Colonna G. Genealogy of an ancient protein family: the Sirtuins, a family of disordered members. BMC Evol Biol 2013; 13:60. [PMID: 23497088 PMCID: PMC3599600 DOI: 10.1186/1471-2148-13-60] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 02/25/2013] [Indexed: 12/21/2022] Open
Abstract
Background Sirtuins genes are widely distributed by evolution and have been found in eubacteria, archaea and eukaryotes. While prokaryotic and archeal species usually have one or two sirtuin homologs, in humans as well as in eukaryotes we found multiple versions and in mammals this family is comprised of seven different homologous proteins being all NAD-dependent de-acylases. 3D structures of human SIRT2, SIRT3, and SIRT5 revealed the overall conformation of the conserved core domain but they were unable to give a structural information about the presence of very flexible and dynamically disordered regions, the role of which is still structurally and functionally unclear. Recently, we modeled the 3D-structure of human SIRT1, the most studied member of this family, that unexpectedly emerged as a member of the intrinsically disordered proteins with its long disordered terminal arms. Despite clear similarities in catalytic cores between the human sirtuins little is known of the general structural characteristics of these proteins. The presence of disorder in human SIRT1 and the propensity of these proteins in promoting molecular interactions make it important to understand the underlying mechanisms of molecular recognition that reasonably should involve terminal segments. The mechanism of recognition, in turn, is a prerequisite for the understanding of any functional activity. Aim of this work is to understand what structural properties are shared among members of this family in humans as well as in other organisms. Results We have studied the distribution of the structural features of N- and C-terminal segments of sirtuins in all known organisms to draw their evolutionary histories by taking into account average length of terminal segments, amino acid composition, intrinsic disorder, presence of charged stretches, presence of putative phosphorylation sites, flexibility, and GC content of genes. Finally, we have carried out a comprehensive analysis of the putative phosphorylation sites in human sirtuins confirming those sites already known experimentally for human SIRT1 and 2 as well as extending their topology to all the family to get feedback of their physiological functions and cellular localization. Conclusions Our results highlight that the terminal segments of the majority of sirtuins possess a number of structural features and chemical and physical properties that strongly support their involvement in activities of recognition and interaction with other protein molecules. We also suggest how a multisite phosphorylation provides a possible mechanism by which flexible and intrinsically disordered segments of a sirtuin supported by the presence of positively or negatively charged stretches might enhance the strength and specificity of interaction with a particular molecular partner.
Collapse
Affiliation(s)
- Susan Costantini
- "Pascale Foundation" National Cancer Institute - Cancer Research Center (CROM), via Ammiraglio Bianco, 83013, Mercogliano, Italy.
| | | | | | | | | | | |
Collapse
|
42
|
Hsu HC, Wang CL, Wang M, Yang N, Chen Z, Sternglanz R, Xu RM. Structural basis for allosteric stimulation of Sir2 activity by Sir4 binding. Genes Dev 2013; 27:64-73. [PMID: 23307867 DOI: 10.1101/gad.208140.112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The budding yeast Sir2 (silent information regulator 2) protein is the founding member of the sirtuin family of NAD-dependent histone/protein deacetylases. Its function in transcriptional silencing requires both the highly conserved catalytic domain and a poorly understood N-terminal regulatory domain (Sir2N). We determined the structure of Sir2 in complex with a fragment of Sir4, a component of the transcriptional silencing complex in Saccharomyces cerevisiae. The structure shows that Sir4 is anchored to Sir2N and contacts the interface between the Sir2N and the catalytic domains through a long loop. We discovered that the interaction between the Sir4 loop and the interdomain interface in Sir2 is critical for allosteric stimulation of the deacetylase activity of Sir2. These results bring to light the structure and function of the regulatory domain of Sir2, and the knowledge should be useful for understanding allosteric regulation of sirtuins in general.
Collapse
Affiliation(s)
- Hao-Chi Hsu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Sirtuins are a family of proteins with NAD(+)-dependent deacetylase or mono-ADP-ribosyltransferase activity. SIRT1, the mammalian ortholog most closely related to Sir2 (the first gene of this family discovered in yeast), exhibits anti-senescence activity in a wide range of mammalian cells. Here, we describe the use of an ex vivo senescence model to study SIRT1 function in primary endothelial cells isolated from the porcine aorta. The methods can be applied to the investigation of the role of SIRT1 in the development of endothelial senescence and atherosclerosis.
Collapse
Affiliation(s)
- Bo Bai
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
44
|
Manjulatha K, Srinivas S, Mulakayala N, Rambabu D, Prabhakar M, Arunasree KM, Alvala M, Basaveswara Rao MV, Pal M. Ethylenediamine diacetate (EDDA) mediated synthesis of aurones under ultrasound: their evaluation as inhibitors of SIRT1. Bioorg Med Chem Lett 2012; 22:6160-5. [PMID: 22929231 DOI: 10.1016/j.bmcl.2012.08.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/29/2012] [Accepted: 08/02/2012] [Indexed: 02/06/2023]
Abstract
An improved synthesis of functionalized aurones has been accomplished via the reaction of benzofuran-3(2H)-one with a range of benzaldehydes in the presence of a mild base EDDA under ultrasound. A number of aurones were synthesized (within 5-30min) and the molecular structure of a representative compound determined by single crystal X-ray diffraction study confirmed Z-geometry of the C-C double bond present within the molecule. Some of the compounds synthesized have shown SIRT1 inhibiting as well as anti proliferative properties against two cancer cell lines in vitro. Compound 3a [(Z)-2-(5-bromo-2-hydroxybenzylidene) benzofuran-3(2H)-one] was identified as a potent inhibitor of SIRT1 (IC(50)=1μM) which showed a dose dependent increase in the acetylation of p53 resulting in induction of apoptosis.
Collapse
Affiliation(s)
- Khanapur Manjulatha
- Institute of Life Sciences, University of Hyderabad Campus, Hyderabad 500 046, India
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pulla VK, Battu MB, Alvala M, Sriram D, Yogeeswari P. Can targeting SIRT-1 to treat type 2 diabetes be a good strategy? A review. Expert Opin Ther Targets 2012; 16:819-32. [PMID: 22762724 DOI: 10.1517/14728222.2012.703656] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Dysregulation of metabolic pathways, caused by imbalances in energy homeostasis, leads to type 2 diabetes characterized by high glucose concentration in the blood due to insulin resistance which is a major disorder in developed countries. AREAS COVERED One of the recent treatment strategies is using activators of SIRT1, which has been in clinical trials. Many of the cellular processes including insulin secretion, cell cycle, and apoptosis are imperatively regulated by a family of mediators called sirtuins. First known mammalian sirtuin, SIRT1 is a positive regulator of insulin secretion, which triggers glucose uptake and utilization. Since the past decade, a major outstanding question is whether SIRT1 activation is a safe therapy for human diseases such as type 2 diabetes? This review summarizes and discusses the advances of the past decade and the challenges that will brazen out perplexity about homeostasis and metabolic pathways linked to SIRT1 and type 2 diabetes. Furthermore, we described the interlink between SIRT1 metabolic pathways of various tissues such as pancreas, skeletal muscle, adipose tissue and liver. EXPERT OPINION However be the complexity of the pathways involved, T2DM regulated by SIRT1 affected metabolism is dropping down progressively due to profound research. In the context of interlinking all the SIRT1 pathways in T2DM we found various crucial intermediaries in metabolic tissues, which can also be targeted for future prospects.
Collapse
Affiliation(s)
- Venkat Koushik Pulla
- Birla Institute of Technology & Science- Pilani, Hyderabad Campus, Department of Pharmacy, Drug Discovery Research Laboratory, R.R. District-500078, Andhra Pradesh, India
| | | | | | | | | |
Collapse
|
46
|
Palladino P, Portella L, Colonna G, Raucci R, Saviano G, Rossi F, Napolitano M, Scala S, Castello G, Costantini S. The N-terminal Region of CXCL11 as Structural Template for CXCR3 Molecular Recognition: Synthesis, Conformational Analysis, and Binding Studies. Chem Biol Drug Des 2012; 80:254-65. [DOI: 10.1111/j.1747-0285.2012.01397.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Pistritto G, Papaleo V, Sanchez P, Ceci C, Barbaccia ML. Divergent modulation of neuronal differentiation by caspase-2 and -9. PLoS One 2012; 7:e36002. [PMID: 22629307 PMCID: PMC3356362 DOI: 10.1371/journal.pone.0036002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 03/27/2012] [Indexed: 11/18/2022] Open
Abstract
Human Ntera2/cl.D1 (NT2) cells treated with retinoic acid (RA) differentiate towards a well characterized neuronal phenotype sharing many features with human fetal neurons. In view of the emerging role of caspases in murine stem cell/neural precursor differentiation, caspases activity was evaluated during RA differentiation. Caspase-2, -3 and -9 activity was transiently and selectively increased in differentiating and non-apoptotic NT2-cells. SiRNA-mediated selective silencing of either caspase-2 (si-Casp2) or -9 (si-Casp9) was implemented in order to dissect the role of distinct caspases. The RA-induced expression of neuronal markers, i.e. neural cell adhesion molecule (NCAM), microtubule associated protein-2 (MAP2) and tyrosine hydroxylase (TH) mRNAs and proteins, was decreased in si-Casp9, but markedly increased in si-Casp2 cells. During RA-induced NT2 differentiation, the class III histone deacetylase Sirt1, a putative caspase substrate implicated in the regulation of the proneural bHLH MASH1 gene expression, was cleaved to a ∼100 kDa fragment. Sirt1 cleavage was markedly reduced in si-Casp9 cells, even though caspase-3 was normally activated, but was not affected (still cleaved) in si-Casp2 cells, despite a marked reduction of caspase-3 activity. The expression of MASH1 mRNA was higher and occurred earlier in si-Casp2 cells, while was reduced at early time points during differentiation in si-Casp9 cells. Thus, caspase-2 and -9 may perform opposite functions during RA-induced NT2 neuronal differentiation. While caspase-9 activation is relevant for proper neuronal differentiation, likely through the fine tuning of Sirt1 function, caspase-2 activation appears to hinder the RA-induced neuronal differentiation of NT2 cells.
Collapse
Affiliation(s)
- Giuseppa Pistritto
- Department of Neuroscience, University of Rome Tor Vergata-Medical School, Rome, Italy.
| | | | | | | | | |
Collapse
|
48
|
Sharma A, Gautam V, Costantini S, Paladino A, Colonna G. Interactomic and pharmacological insights on human sirt-1. Front Pharmacol 2012; 3:40. [PMID: 22470339 PMCID: PMC3311038 DOI: 10.3389/fphar.2012.00040] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 02/23/2012] [Indexed: 12/31/2022] Open
Abstract
Sirt-1 is defined as a nuclear protein involved in the molecular mechanisms of inflammation and neurodegeneration through the de-acetylation of many different substrates even if experimental data in mouse suggest both its cytoplasmatic presence and nucleo-cytoplasmic shuttling upon oxidative stress. Since the experimental structure of human Sirt-1 has not yet been reported, we have modeled its 3D structure, highlighted that it is composed by four different structural regions: N-terminal region, allosteric site, catalytic core and C-terminal region, and underlined that the two terminal regions have high intrinsic disorder propensity and numerous putative phosphorylation sites. Many different papers report experimental studies related to its functional activators because Sirt-1 is implicated in various diseases and cancers. The aim of this article is (i) to present interactomic studies based human Sirt-1 to understand its most important functional relationships in the light of the gene–protein interactions that control major metabolic pathways and (ii) to show by docking studies how this protein binds some activator molecules in order to evidence structural determinants, physico-chemical features and those residues involved in the formation of complexes.
Collapse
Affiliation(s)
- Ankush Sharma
- Research Center of Computational and Biotechnological Sciences, Second University of Naples Naples, Italy
| | | | | | | | | |
Collapse
|
49
|
Alvala M, Bhatnagar S, Ravi A, Jeankumar VU, Manjashetty TH, Yogeeswari P, Sriram D. Novel acridinedione derivatives: design, synthesis, SIRT1 enzyme and tumor cell growth inhibition studies. Bioorg Med Chem Lett 2012; 22:3256-60. [PMID: 22464458 DOI: 10.1016/j.bmcl.2012.03.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/27/2012] [Accepted: 03/07/2012] [Indexed: 10/28/2022]
Abstract
A new scaffold N-(9-(ortho/meta/para-(benzyloxy)phenyl)-3,3,6,6-tetramethyl-1,8-dioxo-1,2,3,4,5,6,7,8-octahydroacridin-10(9H)-yl) isonicotinamide (H1-3) was discovered as a hSIRT1 inhibitor through virtual screening of in-house database. Based on these hits, a library of compounds were designed, synthesized and tested for in vitro hSIRT1 activity. The most potent compound 4d in the series showed a significant inhibition of SIRT1 activity. Further antitumor studies of compound 4d, showed a dose dependent increase in acetylation of p53K382 and decrease in SIRT1 with an IC(50) of 0.25 μM in MDA-MB231 breast cancer cell lines. Individual 3D-QSAR analysis using Schrödinger showed distribution of hydrophobic and non polar positive co-efficient at ortho position essential for bioactivity based on 4d.
Collapse
Affiliation(s)
- Mallika Alvala
- Drug Discovery Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Hyderabad 500 078, India
| | | | | | | | | | | | | |
Collapse
|
50
|
Villalba JM, de Cabo R, Alcain FJ. A patent review of sirtuin activators: an update. Expert Opin Ther Pat 2012; 22:355-67. [PMID: 22475539 DOI: 10.1517/13543776.2012.669374] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Reversible acetylation is a key post-translational modification of target proteins. Sirtuin deacetylases represent the homolog of the yeast silent information regulator (SIR2). Although seven sirtuins have been found in mammals, all sirtuin activators described to date act through SIRT1. AREAS COVERED Areas covered in this paper include a review of the patent literature associated with SIRT1 activators, with a focus on therapeutic applications, primarily related to the use of pharmaceuticals and nutraceuticals containing resveratrol (RSV), and the development of second-generation activators unrelated to RSV. Also discussed is the current controversy over whether or not these molecules are actual SIRT1 activators. EXPERT OPINION Developing effective strategies to protect against diet-induced metabolic imbalance is necessary to fight against current obesity rates. The hypothalamus is a candidate for developing drugs that suppress SIRT1 degradation, as a strategy for treating metabolic syndrome. Deciphering the basic mechanism of activators is essential to develop effective strategies to alter sirtuin activity. This is true regardless of the apparent controversy of whether in vitro activation of SIRT1 is direct or not, depending on the experimental design, and whether sirtuins may play a major role in longevity. The numerous studies on their positive effects against age-related diseases, obesity and other metabolic disorders are still valid, promising to positively influence the development of treatments to improve human health.
Collapse
Affiliation(s)
- José M Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Ciencias, Campus Universitario de Rabanales, Universidad de Córdoba, Spain
| | | | | |
Collapse
|