1
|
Yu H, Li X, Ning B, Feng L, Ren Y, Li S, Kang Y, Ma J, Zhao M. SIRT1: A Potential Therapeutic Target for Coronary Heart Disease Combined with Anxiety or Depression. J Drug Target 2024:1-27. [PMID: 39470049 DOI: 10.1080/1061186x.2024.2422882] [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: 07/08/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Coronary heart disease (CHD) combined with anxiety or depression is increasingly receiving attention in the clinical field of cardiology, and exploring the comorbidity pathological mechanisms of cardiovascular disease combined with psychological disorders is a hot research topic for scholars in this field. Current research suggests that Silent Information Regulatory Factor 1 (SIRT1) may serve as a potential biomarker for the comorbidity mechanism and treatment of CHD with anxiety or depression. SIRT1 is considered a promising therapeutic target for CHD combined with anxiety or depression, with the ability to regulate inflammatory cytokine levels, alleviate oxidative stress damage, activate multiple signaling pathways, reduce platelet hyperresponsiveness, and exert neuroprotective and cardioprotective effects. In this comprehensive review, we deeply studied the structure, function, and mechanism of SIRT1, and discussed its protective effects in the cardiovascular and nervous system. The latest progress in the mechanism of SIRT1's role in CHD combined with anxiety or depression was emphasized, including its specific mechanisms in regulating inflammatory response, alleviating oxidative stress, and mediating various signaling pathways. In addition, this article also summarizes the therapeutic potential of SIRT1 as a potential biomarker in patients with CHD combined with anxiety or depression.
Collapse
Affiliation(s)
- Hubin Yu
- School of Graduate, Shaanxi University of Chinese Medicine, No.1 Middle Section of Shiji Avenue, Xianyang 712046, China
| | - Xinping Li
- School of Graduate, Shaanxi University of Chinese Medicine, No.1 Middle Section of Shiji Avenue, Xianyang 712046, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, No.1 Middle Section of Shiji Avenue, Xianyang 712046, China
| | - Lanshuan Feng
- School of Graduate, Shaanxi University of Chinese Medicine, No.1 Middle Section of Shiji Avenue, Xianyang 712046, China
| | - Yaolong Ren
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Deputy 2, Weiyang West Road, Weicheng District, Xianyang 712000, China
| | - Shilin Li
- School of Graduate, Shaanxi University of Chinese Medicine, No.1 Middle Section of Shiji Avenue, Xianyang 712046, China
| | - Yalong Kang
- School of Graduate, Shaanxi University of Chinese Medicine, No.1 Middle Section of Shiji Avenue, Xianyang 712046, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Air Force Military Medical University, No.15 Changle West Road, Xi'an 710032, China
| | - Mingjun Zhao
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Deputy 2, Weiyang West Road, Weicheng District, Xianyang 712000, China
| |
Collapse
|
2
|
Zhang X, Zhou Y, Liu Y, Li B, Tian S, Zhang Z. Research Progress on the Mechanism and Function of Histone Acetylation Regulating the Interaction between Pathogenic Fungi and Plant Hosts. J Fungi (Basel) 2024; 10:522. [PMID: 39194848 DOI: 10.3390/jof10080522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Histone acetylation is a crucial epigenetic modification, one that holds the key to regulating gene expression by meticulously modulating the conformation of chromatin. Most histone acetylation enzymes (HATs) and deacetylation enzymes (HDACs) in fungi were originally discovered in yeast. The functions and mechanisms of HATs and HDACs in yeast that have been documented offer us an excellent entry point for gaining insights into these two types of enzymes. In the interaction between plants and pathogenic fungi, histone acetylation assumes a critical role, governing fungal pathogenicity and plant immunity. This review paper delves deep into the recent advancements in understanding how histone acetylation shapes the interaction between plants and fungi. It explores how this epigenetic modification influences the intricate balance of power between these two kingdoms of life, highlighting the intricate network of interactions and the subtle shifts in these interactions that can lead to either mutual coexistence or hostile confrontation.
Collapse
Affiliation(s)
- Xiaokang Zhang
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuzhu Zhou
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangzhi Liu
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Boqiang Li
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shiping Tian
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhanquan Zhang
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
3
|
Perico L, Remuzzi G, Benigni A. Sirtuins in kidney health and disease. Nat Rev Nephrol 2024; 20:313-329. [PMID: 38321168 DOI: 10.1038/s41581-024-00806-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2024] [Indexed: 02/08/2024]
Abstract
Sirtuins (SIRTs) are putative regulators of lifespan in model organisms. Since the initial discovery that SIRTs could promote longevity in nematodes and flies, the identification of additional properties of these proteins has led to understanding of their roles as exquisite sensors that link metabolic activity to oxidative states. SIRTs have major roles in biological processes that are important in kidney development and physiological functions, including mitochondrial metabolism, oxidative stress, autophagy, DNA repair and inflammation. Furthermore, altered SIRT activity has been implicated in the pathophysiology and progression of acute and chronic kidney diseases, including acute kidney injury, diabetic kidney disease, chronic kidney disease, polycystic kidney disease, autoimmune diseases and renal ageing. The renoprotective roles of SIRTs in these diseases make them attractive therapeutic targets. A number of SIRT-activating compounds have shown beneficial effects in kidney disease models; however, further research is needed to identify novel SIRT-targeting strategies with the potential to treat and/or prevent the progression of kidney diseases and increase the average human healthspan.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| |
Collapse
|
4
|
Chandrasekaran K, Najimi N, Sagi AR, Yarlagadda S, Salimian M, Arvas MI, Hedayat AF, Kevas Y, Kadakia A, Kristian T, Russell JW. NAD + Precursors Reverse Experimental Diabetic Neuropathy in Mice. Int J Mol Sci 2024; 25:1102. [PMID: 38256175 PMCID: PMC10816262 DOI: 10.3390/ijms25021102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Abnormal NAD+ signaling has been implicated in axonal degeneration in diabetic peripheral neuropathy (DPN). We hypothesized that supplementing NAD+ precursors could alleviate DPN symptoms through increasing the NAD+ levels and activating the sirtuin-1 (SIRT1) protein. To test this, we exposed cultured Dorsal Root Ganglion neurons (DRGs) to Nicotinamide Riboside (NR) or Nicotinamide Mononucleotide (NMN), which increased the levels of NAD+, the SIRT1 protein, and the deacetylation activity that is associated with increased neurite growth. A SIRT1 inhibitor blocked the neurite growth induced via NR or NMN. We then induced neuropathy in C57BL6 mice with streptozotocin (STZ) or a high fat diet (HFD) and administered NR or NMN for two months. Both the STZ and HFD mice developed neuropathy, which was reversed through the NR or NMN administration: sensory function improved, nerve conduction velocities normalized, and intraepidermal nerve fibers were restored. The NAD+ levels and SIRT1 activity were reduced in the DRGs from diabetic mice but were preserved with the NR or NMN treatment. We also tested the effect of NR or NMN administration in mice that overexpress the SIRT1 protein in neurons (nSIRT1 OE) and found no additional benefit from the addition of the drug. These findings suggest that supplementing with NAD+ precursors or activating SIRT1 may be a promising treatment for DPN.
Collapse
Affiliation(s)
- Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Neda Najimi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Avinash R. Sagi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Sushuma Yarlagadda
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Mohammad Salimian
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Muhammed Ikbal Arvas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Ahmad F. Hedayat
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Anand Kadakia
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - James W. Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- CAMC Institute for Academic Medicine, 415 Morris Street Suite 300, Charleston, WV 25301, USA
| |
Collapse
|
5
|
Hu C, Feng Y, Huang G, Cui K, Fan M, Xiang W, Shi Y, Ye D, Ye H, Bai X, Xu F, Xu Y, Huang J. Melatonin prevents EAAC1 deletion-induced retinal ganglion cell degeneration by inhibiting apoptosis and senescence. J Pineal Res 2024; 76:e12916. [PMID: 37786968 DOI: 10.1111/jpi.12916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 10/04/2023]
Abstract
Normal tension glaucoma (NTG) is referred to as a progressive degenerative disorder of the retinal ganglion cells (RGCs), resulting in nonreversible visual defects, despite intraocular pressure levels within the statistically normal range. Current therapeutic strategies for NTG yield limited benefits. Excitatory amino acid carrier 1 (EAAC1) knockout (EAAC1-/- ) in mice has been shown to induce RGC degeneration without elevating intraocular pressure, mimicking pathological characteristics of NTG. In this study, we explored whether daily oral administration of melatonin could block RGCs loss and prevent retinal morphology and function defects associated with EAAC1 deletion. We also explored the molecular mechanisms underlying EAAC1 deletion-induced RGC degeneration and the neuroprotective effects of melatonin. Our RNA sequencing and in vivo data indicated EAAC1 deletion caused elevated oxidative stress, activation of apoptosis and cellular senescence pathways, and neuroinflammation in RGCs. However, melatonin administration efficiently prevented these detrimental effects. Furthermore, we investigated the potential role of apoptosis- and senescence-related redox-sensitive factors in EAAC1 deletion-induced RGCs degeneration and the neuroprotective effects of melatonin administration. We observed remarkable upregulation of p53, whereas NRF2 and Sirt1 expression were significantly decreased in EAAC1-/- mice, which were prevented by melatonin treatment, suggesting that melatonin exerted its neuroprotective effects possibly through modulating NRF2/p53/Sirt1 redox-sensitive signaling pathways. Overall, our study provided a solid foundation for the application of melatonin in the management of NTG.
Collapse
Affiliation(s)
- Chenyang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanlin Feng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Guangyi Huang
- Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Wu Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Huiwen Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Bai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fan Xu
- Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
6
|
Jiang N, Li W, Jiang S, Xie M, Liu R. Acetylation in pathogenesis: Revealing emerging mechanisms and therapeutic prospects. Biomed Pharmacother 2023; 167:115519. [PMID: 37729729 DOI: 10.1016/j.biopha.2023.115519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Protein acetylation modifications play a central and pivotal role in a myriad of biological processes, spanning cellular metabolism, proliferation, differentiation, apoptosis, and beyond, by effectively reshaping protein structure and function. The metabolic state of cells is intricately connected to epigenetic modifications, which in turn influence chromatin status and gene expression patterns. Notably, pathological alterations in protein acetylation modifications are frequently observed in diseases such as metabolic syndrome, cardiovascular disorders, and cancer. Such abnormalities can result in altered protein properties and loss of function, which are closely associated with developing and progressing related diseases. In recent years, the advancement of precision medicine has highlighted the potential value of protein acetylation in disease diagnosis, treatment, and prevention. This review includes provocative and thought-provoking papers outlining recent breakthroughs in acetylation modifications as they relate to cardiovascular disease, mitochondrial metabolic regulation, liver health, neurological health, obesity, diabetes, and cancer. Additionally, it covers the molecular mechanisms and research challenges in understanding the role of acetylation in disease regulation. By summarizing novel targets and prognostic markers for the treatment of related diseases, we aim to contribute to the field. Furthermore, we discuss current hot topics in acetylation research related to health regulation, including N4-acetylcytidine and liquid-liquid phase separation. The primary objective of this review is to provide insights into the functional diversity and underlying mechanisms by which acetylation regulates proteins in disease contexts.
Collapse
Affiliation(s)
- Nan Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Wenyong Li
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Shuanglin Jiang
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Ming Xie
- North China Petroleum Bureau General Hospital, Renqiu 062550, China.
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
7
|
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: 2.0] [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
|
8
|
Razick DI, Akhtar M, Wen J, Alam M, Dean N, Karabala M, Ansari U, Ansari Z, Tabaie E, Siddiqui S. The Role of Sirtuin 1 (SIRT1) in Neurodegeneration. Cureus 2023; 15:e40463. [PMID: 37456463 PMCID: PMC10349546 DOI: 10.7759/cureus.40463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Sirtuins (SIRT) are a class of histone deacetylases that regulate important metabolic pathways and play a role in several disease processes. Of the seven mammalian homologs currently identified, sirtuin 1 (SIRT1) is the best understood and most studied. It has been associated with several neurodegenerative diseases and cancers. As such, it has been further investigated as a therapeutic target in the treatment of disorders such as Parkinson's disease (PD), Huntington's disease (HD), and Alzheimer's disease (AD). SIRT1 deacetylates histones such as H1 lysine 26, H3 lysine 9, H3 lysine 56, and H4 lysine 16 to regulate chromatin remodeling and gene transcription. The homolog has also been observed to express contradictory responses to tumor suppression and tumor promotion. Studies have shown that SIRT1 may have anti-inflammatory properties by inhibiting the effects of NF-κB, as well as stimulating upregulation of autophagy. The SIRT1 activators resveratrol and cilostazol have been shown to improve Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) scores in AD patients. In this review, we aim to explore the various roles of SIRT1 with regard to neuroprotection and neurodegeneration.
Collapse
Affiliation(s)
- Daniel I Razick
- Surgery, California Northstate University College of Medicine, Elk Grove, USA
| | - Muzammil Akhtar
- Surgery, California Northstate University College of Medicine, Elk Grove, USA
| | - Jimmy Wen
- Physical Medicine and Rehabilitation, California Northstate University College of Medicine, Elk Grove, USA
| | - Meraj Alam
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Nabeal Dean
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Muhammad Karabala
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Ubaid Ansari
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Zaid Ansari
- Internal Medicine, University of California Berkeley, Berkeley, USA
| | - Ethan Tabaie
- Neurosurgery, California Northstate University College of Medicine, Elk Grove, USA
| | - Shakeel Siddiqui
- Anesthesiology, OrthoMed Staffing Anesthesiology Group, Dallas, USA
| |
Collapse
|
9
|
Zessin M, Meleshin M, Hilscher S, Schiene-Fischer C, Barinka C, Jung M, Schutkowski M. Continuous Fluorescent Sirtuin Activity Assay Based on Fatty Acylated Lysines. Int J Mol Sci 2023; 24:ijms24087416. [PMID: 37108579 PMCID: PMC10138348 DOI: 10.3390/ijms24087416] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Lysine deacetylases, like histone deacetylases (HDACs) and sirtuins (SIRTs), are involved in many regulatory processes such as control of metabolic pathways, DNA repair, and stress responses. Besides robust deacetylase activity, sirtuin isoforms SIRT2 and SIRT3 also show demyristoylase activity. Interestingly, most of the inhibitors described so far for SIRT2 are not active if myristoylated substrates are used. Activity assays with myristoylated substrates are either complex because of coupling to enzymatic reactions or time-consuming because of discontinuous assay formats. Here we describe sirtuin substrates enabling direct recording of fluorescence changes in a continuous format. Fluorescence of the fatty acylated substrate is different when compared to the deacylated peptide product. Additionally, the dynamic range of the assay could be improved by the addition of bovine serum albumin, which binds the fatty acylated substrate and quenches its fluorescence. The main advantage of the developed activity assay is the native myristoyl residue at the lysine side chain avoiding artifacts resulting from the modified fatty acyl residues used so far for direct fluorescence-based assays. Due to the extraordinary kinetic constants of the new substrates (KM values in the low nM range, specificity constants between 175,000 and 697,000 M-1s-1) it was possible to reliably determine the IC50 and Ki values for different inhibitors in the presence of only 50 pM of SIRT2 using different microtiter plate formats.
Collapse
Affiliation(s)
- Matthes Zessin
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Marat Meleshin
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Sebastian Hilscher
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Cordelia Schiene-Fischer
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Cyril Barinka
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, 25250 Vestec, Czech Republic
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Mike Schutkowski
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| |
Collapse
|
10
|
Navarrete B, Ibeas JI, Barrales RR. Systematic characterization of Ustilago maydis sirtuins shows Sir2 as a modulator of pathogenic gene expression. Front Microbiol 2023; 14:1157990. [PMID: 37113216 PMCID: PMC10126416 DOI: 10.3389/fmicb.2023.1157990] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Phytopathogenic fungi must adapt to the different environmental conditions found during infection and avoid the immune response of the plant. For these adaptations, fungi must tightly control gene expression, allowing sequential changes in transcriptional programs. In addition to transcription factors, chromatin modification is used by eukaryotic cells as a different layer of transcriptional control. Specifically, the acetylation of histones is one of the chromatin modifications with a strong impact on gene expression. Hyperacetylated regions usually correlate with high transcription and hypoacetylated areas with low transcription. Thus, histone deacetylases (HDACs) commonly act as repressors of transcription. One member of the family of HDACs is represented by sirtuins, which are deacetylases dependent on NAD+, and, thus, their activity is considered to be related to the physiological stage of the cells. This property makes sirtuins good regulators during environmental changes. However, only a few examples exist, and with differences in the extent of the implication of the role of sirtuins during fungal phytopathogenesis. In this work, we have performed a systematic study of sirtuins in the maize pathogen Ustilago maydis, finding Sir2 to be involved in the dimorphic switch from yeast cell to filament and pathogenic development. Specifically, the deletion of sir2 promotes filamentation, whereas its overexpression highly reduces tumor formation in the plant. Moreover, transcriptomic analysis revealed that Sir2 represses genes that are expressed during biotrophism development. Interestingly, our results suggest that this repressive effect is not through histone deacetylation, indicating a different target of Sir2 in this fungus.
Collapse
|
11
|
Yin JY, Lu XT, Hou ML, Cao T, Tian Z. Sirtuin1-p53: a potential axis for cancer therapy. Biochem Pharmacol 2023; 212:115543. [PMID: 37037265 DOI: 10.1016/j.bcp.2023.115543] [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: 11/26/2022] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/12/2023]
Abstract
Sirtuin1 (SIRT1) is a conserved nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase that plays key roles in a range of cellular events, including the maintenance of genome stability, gene regulation, cell proliferation, and apoptosis. P53 is one of the most studied tumor suppressors and the first identified non-histone target of SIRT1. SIRT1 deacetylates p53 in a NAD+-dependent manner and inhibits its transcriptional activity, thus exerting action on a series of pathways related to tissue homeostasis and various pathological states. The SIRT1-p53 axis is thought to play a central role in tumorigenesis. Although SIRT1 was initially identified as a tumor promoter, evidence now indicates that SIRT1 may also act as a tumor suppressor. This seemingly contradictory evidence indicates that the functionality of SIRT1 may be dictated by different cell types and intracellular localization patterns. In this review, we summarize recent evidence relating to the interactions between SIRT1 and p53 and discuss the relative roles of these two molecules with regards to cancer-associated cellular events. We also provide an overview of current knowledge of SIRT1-p53 signaling in tumorigenesis. Given the vital role of the SIRT1-p53 pathway, targeting this axis may provide promising strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Jia-Yi Yin
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xin-Tong Lu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Meng-Ling Hou
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Ting Cao
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Zhen Tian
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| |
Collapse
|
12
|
Xiao B, Li D, Liao B, Zheng H, Yang X, Xie Y, Xie Z, Li C. Effects of microplastic combined with Cr(III) on apoptosis and energy pathway of coral endosymbiont. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:39750-39763. [PMID: 36602726 DOI: 10.1007/s11356-022-25041-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 12/24/2022] [Indexed: 06/17/2023]
Abstract
The combined effect of polyethylene (PE) microplastics and chromium (Cr(III)) on the scleractinian coral Acropora pruinosa (A. pruinosa) was investigated. The endpoints analysed in this study included the endosymbiont density, the chlorophyll a + c content, and the activity of enzymes involved in apoptosis (caspase-1, caspase-3), glycolysis (lactate dehydrogenase, LDH), the pentose phosphate pathway (glucose-6-phosphate dehydrogenase, G6PDH) and electron transfer coenzyme (nicotinamide adenine dinucleotide, NAD+/NADH). During the 7-day exposure to PE and Cr(III) stress, the endosymbiont density and chlorophyll content decreased gradually. The caspase-1 and caspase-3 activities increased in the high-concentration Cr(III) exposure group. Furthermore, the LDH and G6PDH activities decreased significantly, and the NAD+/NADH was decreased significantly. In summary, the results showed that PE and Cr(III) stress inhibited the endosymbiont energy metabolism enzymes and further led to endosymbiont apoptosis in coral. In addition, under exposure to the combination of stressors, when the concentration of Cr(III) remained at 1 × 10-2 mg/L, the toxic effects of heavy metals on the endosymbiont were temporarily relieved with elevated PE concentrations. In contrast, when coral polyps were exposed to 5 mg/L PE and increasing Cr(III) concentrations, their metabolic activities were seriously disturbed, which increased the burden of energy consumption. In the short term, the toxic effect of Cr(III) was more obvious than that of PE because Cr(III) exposure leads to endosymbiont apoptosis and irreversible damage. This is the first study to provide insights into the combined effect of microplastic and Cr(III) stress on the apoptosis and energy pathways of coral endosymbionts. This study suggested that microplastics combined with Cr(III) are an important factor affecting the apoptosis and energy metabolism of endosymbionts, accelerating the collapse of the balance between the coral host and symbiotic endosymbiont.
Collapse
Affiliation(s)
- Baohua Xiao
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Dongdong Li
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Baolin Liao
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Huina Zheng
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Xiaodong Yang
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Yongqi Xie
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Ziqiang Xie
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China
| | - Chengyong Li
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, People's Republic of China.
- School of Chemistry and Environment, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, People's Republic of China.
| |
Collapse
|
13
|
Focus on the Use of Resveratrol in Bladder Cancer. Int J Mol Sci 2023; 24:ijms24054562. [PMID: 36901993 PMCID: PMC10003096 DOI: 10.3390/ijms24054562] [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: 01/17/2023] [Revised: 02/13/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Bladder cancer is the most common tumor of the urinary system, with a high incidence in the male population. Surgery and intravesical instillations can eradicate it, although recurrences are very common, with possible progression. For this reason, adjuvant therapy should be considered in all patients. Resveratrol displays a biphasic dose response both in vitro and in vivo (intravesical application) with an antiproliferative effect at high concentrations and antiangiogenic action in vivo (intraperitoneal application) at a low concentration, suggesting a potential role for it in clinical management as an adjuvant to conventional therapy. In this review, we examine the standard therapeutical approach to bladder cancer and the preclinical studies that have investigated resveratrol in xenotransplantation models of bladder cancer. Molecular signals are also discussed, with a particular focus on the STAT3 pathway and angiogenic growth factor modulation.
Collapse
|
14
|
Smirnov D, Eremenko E, Stein D, Kaluski S, Jasinska W, Cosentino C, Martinez-Pastor B, Brotman Y, Mostoslavsky R, Khrameeva E, Toiber D. SIRT6 is a key regulator of mitochondrial function in the brain. Cell Death Dis 2023; 14:35. [PMID: 36653345 PMCID: PMC9849342 DOI: 10.1038/s41419-022-05542-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
The SIRT6 deacetylase has been implicated in DNA repair, telomere maintenance, glucose and lipid metabolism and, importantly, it has critical roles in the brain ranging from its development to neurodegeneration. Here, we combined transcriptomics and metabolomics approaches to characterize the functions of SIRT6 in mouse brains. Our analysis reveals that SIRT6 is a central regulator of mitochondrial activity in the brain. SIRT6 deficiency in the brain leads to mitochondrial deficiency with a global downregulation of mitochondria-related genes and pronounced changes in metabolite content. We suggest that SIRT6 affects mitochondrial functions through its interaction with the transcription factor YY1 that, together, regulate mitochondrial gene expression. Moreover, SIRT6 target genes include SIRT3 and SIRT4, which are significantly downregulated in SIRT6-deficient brains. Our results demonstrate that the lack of SIRT6 leads to decreased mitochondrial gene expression and metabolomic changes of TCA cycle byproducts, including increased ROS production, reduced mitochondrial number, and impaired membrane potential that can be partially rescued by restoring SIRT3 and SIRT4 levels. Importantly, the changes we observed in SIRT6-deficient brains are also occurring in aging human brains and particularly in patients with Alzheimer's, Parkinson's, Huntington's, and Amyotrophic lateral sclerosis disease. Overall, our results suggest that the reduced levels of SIRT6 in the aging brain and neurodegeneration initiate mitochondrial dysfunction by altering gene expression, ROS production, and mitochondrial decay.
Collapse
Affiliation(s)
- Dmitrii Smirnov
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | - Ekaterina Eremenko
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Daniel Stein
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Shai Kaluski
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Weronika Jasinska
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Claudia Cosentino
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Barbara Martinez-Pastor
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), Madrid, 28029, Spain
| | - Yariv Brotman
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Ekaterina Khrameeva
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, 121205, Russia.
| | - Debra Toiber
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel.
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel.
| |
Collapse
|
15
|
Wang K, Liu H, Hu Q, Wang L, Liu J, Zheng Z, Zhang W, Ren J, Zhu F, Liu GH. Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal Transduct Target Ther 2022; 7:374. [PMID: 36336680 PMCID: PMC9637765 DOI: 10.1038/s41392-022-01211-8] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Aging is accompanied by the decline of organismal functions and a series of prominent hallmarks, including genetic and epigenetic alterations. These aging-associated epigenetic changes include DNA methylation, histone modification, chromatin remodeling, non-coding RNA (ncRNA) regulation, and RNA modification, all of which participate in the regulation of the aging process, and hence contribute to aging-related diseases. Therefore, understanding the epigenetic mechanisms in aging will provide new avenues to develop strategies to delay aging. Indeed, aging interventions based on manipulating epigenetic mechanisms have led to the alleviation of aging or the extension of the lifespan in animal models. Small molecule-based therapies and reprogramming strategies that enable epigenetic rejuvenation have been developed for ameliorating or reversing aging-related conditions. In addition, adopting health-promoting activities, such as caloric restriction, exercise, and calibrating circadian rhythm, has been demonstrated to delay aging. Furthermore, various clinical trials for aging intervention are ongoing, providing more evidence of the safety and efficacy of these therapies. Here, we review recent work on the epigenetic regulation of aging and outline the advances in intervention strategies for aging and age-associated diseases. A better understanding of the critical roles of epigenetics in the aging process will lead to more clinical advances in the prevention of human aging and therapy of aging-related diseases.
Collapse
Affiliation(s)
- Kang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Huicong Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Qinchao Hu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China
- Hospital of Stomatology, Sun Yat-sen University, 510060, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 510060, Guangzhou, China
| | - Lingna Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Jiaqing Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Zikai Zheng
- University of Chinese Academy of Sciences, 100049, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, 100049, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jie Ren
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Fangfang Zhu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
| |
Collapse
|
16
|
Tan S, Li X. Small-Molecule Fluorescent Probes for Detecting HDAC Activity. Chem Asian J 2022; 17:e202200835. [PMID: 36117388 DOI: 10.1002/asia.202200835] [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: 08/11/2022] [Revised: 09/15/2022] [Indexed: 11/05/2022]
Abstract
Histone deacetylases (HDACs) play critical roles in epigenetic modification. These enzymes can remove acetyl groups from the N-terminal lysine residues of histones, thereby regulating gene expression. Because of their great relevance to various diseases, numerous HDAC inhibitors have been developed. In this context, assays for HDAC activity are prerequisite. Due to the advantages of small-molecule fluorescent probes, researchers have developed many probes to detect HDAC activity for developing HDAC inhibitors. Based on the mechanism of action, two main types of small-molecule fluorescent probes are known. One type is based on binding affinity that are generally HDAC inhibitor-fluorophore conjugates. The other one is enzyme-activated probes, which act as HDAC substrates and show fluorogenic or ratiometric response after being deacetylated by HDACs.
Collapse
Affiliation(s)
- Shuyu Tan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xin Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
17
|
Li J, Cao Y, Niu K, Qiu J, Wang H, You Y, Li D, Luo Y, Zhu Z, Zhang Y, Liu N. Quantitative acetylomics reveals dynamics of protein lysine acetylation in mouse livers during aging and upon the treatment of nicotinamide mononucleotide. Mol Cell Proteomics 2022; 21:100276. [PMID: 35931320 PMCID: PMC9436820 DOI: 10.1016/j.mcpro.2022.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/25/2022] [Accepted: 07/27/2022] [Indexed: 10/24/2022] Open
Abstract
Lysine acetylation is a reversible and dynamic post-translational modification that play vital roles in regulating multiple cellular processes including aging. However, acetylome-wide analysis in the aging process remains poorly studied in mammalian tissues. Nicotinamide adenine dinucleotide (NAD+), a hub metabolite, benefits healthspan at least in part due to the activation of Sirtuins, a family of NAD+-consuming deacetylases, indicating changes in acetylome. Here, we combine two antibodies for the enrichment of acetylated peptides and perform label-free quantitative acetylomic analysis of mouse livers during natural aging and upon the treatment of beta-nicotinamide mononucleotide (NMN), a NAD+ booster. Our study describes previously unknown acetylation sites and reveals the acetylome-wide dynamics with age as well as upon the treatment of NMN. We discover protein acetylation events as potential aging biomarkers. We demonstrate that the life-beneficial effect of NMN could be partially reflected by the changes in age-related protein acetylation. Our quantitative assessment indicates that NMN has mild effects on acetylation sites previously reported as substrates of Sirtuins. Collectively, our data analyzes protein acetylation with age, laying critical foundation for the functional study of protein post-translational modification essential for healthy aging and perhaps disease conditions.
Collapse
Affiliation(s)
- Jingshu Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ye Cao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kongyan Niu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaqian Qiu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingnan You
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dean Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu Luo
- Abiochem Biotechnology, 1299 Zi Yue Rd., Shanghai, 200241, China
| | - Zhengjiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China.
| | - Nan Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai, 201210, China.
| |
Collapse
|
18
|
Zhao G, Rusche LN. Sirtuins in Epigenetic Silencing and Control of Gene Expression in Model and Pathogenic Fungi. Annu Rev Microbiol 2022; 76:157-178. [PMID: 35609947 DOI: 10.1146/annurev-micro-041020-100926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fungi, including yeasts, molds, and mushrooms, proliferate on decaying matter and then adopt quiescent forms once nutrients are depleted. This review explores how fungi use sirtuin deacetylases to sense and respond appropriately to changing nutrients. Because sirtuins are NAD+-dependent deacetylases, their activity is sensitive to intracellular NAD+ availability. This allows them to transmit information about a cell's metabolic state on to the biological processes they influence. Fungal sirtuins are primarily known to deacetylate histones, repressing transcription and modulating genome stability. Their target genes include those involved in NAD+ homeostasis, metabolism, sporulation, secondary metabolite production, and virulence traits of pathogenic fungi. By targeting different genes over evolutionary time, sirtuins serve as rewiring points that allow organisms to evolve novel responses to low NAD+ stress by bringing relevant biological processes under the control of sirtuins. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Guolei Zhao
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA; ,
| | - Laura N Rusche
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA; ,
| |
Collapse
|
19
|
Ogle MM, Trevino R, Schell J, Varmazyad M, Horikoshi N, Gius D. Manganese Superoxide Dismutase Acetylation and Regulation of Protein Structure in Breast Cancer Biology and Therapy. Antioxidants (Basel) 2022; 11:635. [PMID: 35453320 PMCID: PMC9024550 DOI: 10.3390/antiox11040635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/01/2023] Open
Abstract
The loss and/or dysregulation of several cellular and mitochondrial antioxidants' expression or enzymatic activity, which leads to the aberrant physiological function of these proteins, has been shown to result in oxidative damage to cellular macromolecules. In this regard, it has been surmised that the disruption of mitochondrial networks responsible for maintaining normal metabolism is an established hallmark of cancer and a novel mechanism of therapy resistance. This altered metabolism leads to aberrant accumulation of reactive oxygen species (ROS), which, under specific physiological conditions, leads to a potential tumor-permissive cellular environment. In this regard, it is becoming increasingly clear that the loss or disruption of mitochondrial oxidant scavenging enzymes may be, in specific tumors, either an early event in transformation or exhibit tumor-promoting properties. One example of such an antioxidant enzyme is manganese superoxide dismutase (MnSOD, also referred to as SOD2), which detoxifies superoxide, a ROS that has been shown, when its normal physiological levels are disrupted, to lead to oncogenicity and therapy resistance. Here, we will also discuss how the acetylation of MnSOD leads to a change in detoxification function that leads to a cellular environment permissive for the development of lineage plasticity-like properties that may be one mechanism leading to tumorigenic and therapy-resistant phenotypes.
Collapse
Affiliation(s)
- Meredith M. Ogle
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; (M.M.O.); (R.T.J.); (J.S.); (M.V.); (N.H.)
- Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Rolando Trevino
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; (M.M.O.); (R.T.J.); (J.S.); (M.V.); (N.H.)
- Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Joseph Schell
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; (M.M.O.); (R.T.J.); (J.S.); (M.V.); (N.H.)
- Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Mahboubeh Varmazyad
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; (M.M.O.); (R.T.J.); (J.S.); (M.V.); (N.H.)
- Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; (M.M.O.); (R.T.J.); (J.S.); (M.V.); (N.H.)
- Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - David Gius
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; (M.M.O.); (R.T.J.); (J.S.); (M.V.); (N.H.)
- Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| |
Collapse
|
20
|
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: 148] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [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
- *Correspondence: Dahai Hu, ; Jun Tie,
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Dahai Hu, ; Jun Tie,
| |
Collapse
|
21
|
Cai Q, Tian L, Xie JT, Jiang DH, Keyhani NO. Contributions of a Histone Deacetylase (SirT2/Hst2) to Beauveria bassiana Growth, Development, and Virulence. J Fungi (Basel) 2022; 8:jof8030236. [PMID: 35330238 PMCID: PMC8950411 DOI: 10.3390/jof8030236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Sirtuins are a class of histone deacetylases that promote heterochromatin formation to repress transcription. The entomopathogenic fungus Beauveria bassiana contains six sirtuin homologs. The class III histone deacetylase, BbSir2, has been previously shown to affect the regulation of carbon/nitrogen metabolism and asexual development, with only moderate effects on virulence. Here, we examine another class III histone deacetylase (BbSirT2) and show that it contributes to deacetylation of lysine residues on histone H4-K16ac. Directed gene-knockout of BbSirT2 dramatically reduced conidiation, the ability of the fungus to metabolize a range of carbon and nitrogen sources, and tolerances to oxidative, heat, and UV stress and significantly attenuated virulence in both intrahemocoel injection and topical bioassays using the Greater wax moth (Galleria mellonella) as the insect host. ΔBbSirT2 cells showed alterations in cell cycle development and hyphal septation and produced morphologically aberrant conidia. Comparative transcriptomic analyses of wild type versus ΔBbSirT2 cells indicated differential expression of 1148 genes. Differentially expressed genes were enriched in pathways involved in cell cycle and rescue, carbon/nitrogen metabolism, and pathogenesis. These included changes in the expression of polyketide synthases (PKSs) and LysM effector proteins that contribute to degradation of host toxins and target host pathways, respectively. These data indicate contributions of BbSirT2 in helping to mediate fungal stress and development, with the identification of affected gene targets that can help account for the observed reduced virulence phenotype.
Collapse
Affiliation(s)
- Qing Cai
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.-T.X.); (D.-H.J.)
- Department of Microbiology and Cell Science, University of Florida, Bldg. 981, Museum Rd., Gainesville, FL 32611, USA
- Correspondence: (Q.C.); (N.O.K.)
| | - Li Tian
- Shandong Provincial Key Laboratory of Microbial Engineering, Department of Bioengineering, Qilu University of Technology, Jinan 250353, China;
| | - Jia-Tao Xie
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.-T.X.); (D.-H.J.)
| | - Dao-Hong Jiang
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.-T.X.); (D.-H.J.)
| | - Nemat O. Keyhani
- Department of Microbiology and Cell Science, University of Florida, Bldg. 981, Museum Rd., Gainesville, FL 32611, USA
- Correspondence: (Q.C.); (N.O.K.)
| |
Collapse
|
22
|
Hrubša M, Siatka T, Nejmanová I, Vopršalová M, Kujovská Krčmová L, Matoušová K, Javorská L, Macáková K, Mercolini L, Remião F, Máťuš M, Mladěnka P. Biological Properties of Vitamins of the B-Complex, Part 1: Vitamins B 1, B 2, B 3, and B 5. Nutrients 2022; 14:484. [PMID: 35276844 PMCID: PMC8839250 DOI: 10.3390/nu14030484] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
This review summarizes the current knowledge on essential vitamins B1, B2, B3, and B5. These B-complex vitamins must be taken from diet, with the exception of vitamin B3, that can also be synthetized from amino acid tryptophan. All of these vitamins are water soluble, which determines their main properties, namely: they are partly lost when food is washed or boiled since they migrate to the water; the requirement of membrane transporters for their permeation into the cells; and their safety since any excess is rapidly eliminated via the kidney. The therapeutic use of B-complex vitamins is mostly limited to hypovitaminoses or similar conditions, but, as they are generally very safe, they have also been examined in other pathological conditions. Nicotinic acid, a form of vitamin B3, is the only exception because it is a known hypolipidemic agent in gram doses. The article also sums up: (i) the current methods for detection of the vitamins of the B-complex in biological fluids; (ii) the food and other sources of these vitamins including the effect of common processing and storage methods on their content; and (iii) their physiological function.
Collapse
Affiliation(s)
- Marcel Hrubša
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (M.H.); (M.V.); (P.M.)
| | - Tomáš Siatka
- Department of Pharmacognosy, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (T.S.); (K.M.)
| | - Iveta Nejmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Marie Vopršalová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (M.H.); (M.V.); (P.M.)
| | - Lenka Kujovská Krčmová
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic;
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Kralove, Czech Republic; (K.M.); (L.J.)
| | - Kateřina Matoušová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Kralove, Czech Republic; (K.M.); (L.J.)
| | - Lenka Javorská
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Kralove, Czech Republic; (K.M.); (L.J.)
| | - Kateřina Macáková
- Department of Pharmacognosy, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (T.S.); (K.M.)
| | - Laura Mercolini
- Research Group of Pharmaco-Toxicological Analysis (PTA Lab), Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Fernando Remião
- UCIBIO—Applied Molecular Biosciences Unit, REQUINTE, Toxicology Laboratory, Biological Sciences Department Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Marek Máťuš
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovak Republic
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (M.H.); (M.V.); (P.M.)
| | | |
Collapse
|
23
|
Avilkina V, Chauveau C, Ghali Mhenni O. Sirtuin function and metabolism: Role in pancreas, liver, and adipose tissue and their crosstalk impacting bone homeostasis. Bone 2022; 154:116232. [PMID: 34678494 DOI: 10.1016/j.bone.2021.116232] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Mammalian sirtuins (SIRT1-7) are members of the nicotine adenine dinucleotide (NAD+)-dependent family of enzymes critical for histone deacetylation and posttranslational modification of proteins. Sirtuin family members regulate a wide spectrum of biological processes and are best known for maintaining longevity. Sirtuins are well characterized in metabolic tissues such as the pancreas, liver and adipose tissue (AT). They are regulated by a diverse range of stimuli, including nutrients and metabolic changes within the organism. Indeed, nutrient-associated conditions, such as obesity and anorexia nervosa (AN), were found to be associated with bone fragility development in osteoporosis. Interestingly, it has also been demonstrated that sirtuins, more specifically SIRT1, can regulate bone activity. Various studies have demonstrated the importance of sirtuins in bone in the regulation of bone homeostasis and maintenance of the balance between bone resorption and bone formation. However, to understand the molecular mechanisms involved in the negative regulation of bone homeostasis during overnutrition (obesity) or undernutrition, it is crucial to examine a wider picture and to determine the pancreatic, liver and adipose tissue pathway crosstalk responsible for bone loss. Particularly, under AN conditions, sirtuin family members are highly expressed in metabolic tissue, but this phenomenon is reversed in bone, and severe bone loss has been observed in human subjects. AN-associated bone loss may be connected to SIRT1 deficiency; however, additional factors may interfere with bone homeostasis. Thus, in this review, we focus on sirtuin activity in the pancreas, liver and AT in cases of over- and undernutrition, especially the regulation of their secretome by sirtuins. Furthermore, we examine how the secretome of the pancreas, liver and AT affects bone homeostasis, focusing on undernutrition. This review aims to lead to a better understanding of the crosstalk between sirtuins, metabolic organs and bone. In long term prospective it should contribute to promote improvement of therapeutic strategies for the prevention of metabolic diseases and the development of osteoporosis.
Collapse
Affiliation(s)
- Viktorija Avilkina
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Univ. Littoral Côte d'Opale, F-62200, Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Univ. Littoral Côte d'Opale, F-62200, Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Olfa Ghali Mhenni
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Univ. Littoral Côte d'Opale, F-62200, Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
24
|
Fujisue K, Yamamoto E, Sueta D, Takae M, Nishihara T, Komorita T, Usuku H, Yamanaga K, Ito M, Hoshiyama T, Kanazawa H, Takashio S, Arima Y, Araki S, Soejima H, Kaikita K, Matsushita K, Tsujita K. Increased soluble programed cell death-ligand 1 is associated with acute coronary syndrome. Int J Cardiol 2021; 349:1-6. [PMID: 34843822 DOI: 10.1016/j.ijcard.2021.11.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/26/2021] [Accepted: 11/24/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Programmed cell death (PD)-1 and its ligand (PD-L1) plays crucial roles in T-cell tolerance as immune checkpoint. Previous studies reported that increased serum levels of soluble PD-L1 (sPD-L1) reflect myocardial and vascular inflammation. However, little is known about the clinical relationship between sPD-L1 and acute coronary syndrome (ACS). We investigated the relation of sPD-L1 and ACS. METHODS We prospectively measured serum levels of sPD-L1 using a commercially available enzyme-linked immunosorbent assay kit in patients with coronary artery disease (CAD) and continuous non-CAD admitted to Kumamoto University Hospital between December 2017 and June 2019. All malignant diseases, patients who underwent hemodialysis, active collagen diseases, and severe infectious diseases were excluded. RESULTS Totally, 446 CAD patients [ACS, n = 124; chronic coronary syndrome (CCS), n = 322] and 24 non-CAD patients were analyzed. The levels of sPD-L1 were significantly higher in patients with ACS than those both with non-CAD and CCS {ACS, 188.7 (111.0-260.8) pg/mL, p < 0.001 vs. non-CAD [83.5 (70.8-130.4) pg/mL]; and p = 0.009 vs. CCS [144.2 (94.8-215.5) pg/mL], respectively}. Univariate logistic regression analysis identified that sPD-L1 was significantly associated with ACS [odds ratio (OR): 1.459, 95% confidence interval (CI): 1.198-1.778, p < 0.001]. Multivariable logistic regression analysis with nine significant factors identified from the univariate analysis revealed that sPD-L1 was significantly and independently associated with ACS (OR: 1.561, 95% CI: 1.215-2.006, p < 0.001). CONCLUSIONS This is the first clinical study to demonstrate the increased level of sPD-L1 in patients with CAD, and the significant association with ACS.
Collapse
Affiliation(s)
- Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan.
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Masafumi Takae
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Taiki Nishihara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Takashi Komorita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Hiroki Usuku
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Kenshi Yamanaga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Tadashi Hoshiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Hirofumi Soejima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Kenichi Matsushita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
25
|
Zessin M, Meleshin M, Simic Z, Kalbas D, Arbach M, Gebhardt P, Melesina J, Liebscher S, Bordusa F, Sippl W, Barinka C, Schutkowski M. Continuous Sirtuin/HDAC (histone deacetylase) activity assay using thioamides as PET (Photoinduced Electron Transfer)-based fluorescence quencher. Bioorg Chem 2021; 117:105425. [PMID: 34695733 DOI: 10.1016/j.bioorg.2021.105425] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/28/2022]
Abstract
Histone deacylase 11 and human sirtuins are able to remove fatty acid-derived acyl moieties from the ε-amino group of lysine residues. Specific substrates are needed for investigating the biological functions of these enzymes. Additionally, appropriate screening systems are required for identification of modulators of enzymatic activities of HDAC11 and sirtuins. We designed and synthesized a set of activity probes by incorporation of a thioamide quencher unit into the fatty acid-derived acyl chain and a fluorophore in the peptide sequence. Systematic variation of both fluorophore and quencher position resulted "super-substrates" with catalytic constants of up to 15,000,000 M-1s-1 for human sirtuin 2 (Sirt2) enabling measurements using enzyme concentrations down to 100 pM in microtiter plate-based screening formats. It could be demonstrated that the stalled intermediate formed by the reaction of Sirt2-bound thiomyristoylated peptide and NAD+ has IC50 values below 200 pM.
Collapse
Affiliation(s)
- Matthes Zessin
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Marat Meleshin
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Zeljko Simic
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Diana Kalbas
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Miriam Arbach
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Philip Gebhardt
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Jelena Melesina
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Sandra Liebscher
- Department of Natural Product Biochemistry, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Frank Bordusa
- Department of Natural Product Biochemistry, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Mike Schutkowski
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany.
| |
Collapse
|
26
|
Abstract
Sirtuin1 is a nutrient-sensitive class III histone deacetylase which is a well-known regulator of organismal lifespan. It has been extensively studied for its role in metabolic regulation as well. Along with its involvement in ageing and metabolism, Sirtuin1 directly deacetylates many critical proteins controlling cardiovascular pathophysiology. Studies using conditional expression and deletion of Sirtuin1 have revealed that it functions in a highly tissue/organ-specific manner. In the vasculature, Sirtuin1 controls endothelial homoeostasis by governing the expression of inflammatory mediators, oxidants and essential transcription factors. Adding to this complexity, Sirtuin1 expression and/or function is also governed by some of these target proteins. Therefore, the importance of better understanding the organ and tissue specificity of Sirtuin1 is highly desirable. Considering the huge volume of research done in this field, this review focuses on Sirtuin1 targets regulating vascular endothelial function. Here, we summarize the discovery of Sirtuin1 as a transcription controller and the further identification of direct target proteins involved in the vascular physiology. Overall, this review presents a holistic picture of the complex cross-talk involved in the molecular regulation of vascular physiology by Sirtuin1.
Collapse
Affiliation(s)
- Jitendra Kumar
- François M. Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Santosh Kumar
- François M. Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
27
|
Jakoube P, Cutano V, González-Morena JM, Keckesova Z. Mitochondrial Tumor Suppressors-The Energetic Enemies of Tumor Progression. Cancer Res 2021; 81:4652-4667. [PMID: 34183354 PMCID: PMC9397617 DOI: 10.1158/0008-5472.can-21-0518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023]
Abstract
Tumor suppressors represent a critical line of defense against tumorigenesis. Their mechanisms of action and the pathways they are involved in provide important insights into cancer progression, vulnerabilities, and treatment options. Although nuclear and cytosolic tumor suppressors have been extensively investigated, relatively little is known about tumor suppressors localized within the mitochondria. However, recent research has begun to uncover the roles of these important proteins in suppressing tumorigenesis. Here, we review this newly developing field and summarize available information on mitochondrial tumor suppressors.
Collapse
Affiliation(s)
- Pavel Jakoube
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Valentina Cutano
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Juan M. González-Morena
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Keckesova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.,Corresponding Author: Zuzana Keckesova, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 16000, Czech Republic. Phone: 420-2201-83584; E-mail:
| |
Collapse
|
28
|
Zheng M, Hu C, Wu M, Chin YE. Emerging role of SIRT2 in non-small cell lung cancer. Oncol Lett 2021; 22:731. [PMID: 34429771 PMCID: PMC8371967 DOI: 10.3892/ol.2021.12992] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 04/16/2021] [Indexed: 11/14/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most devastating cancer types, accounting for >80% of lung cancer cases. The median relative survival time of patients with NSCLC is <1 year. Lysine acetylation is a major post-translational modification that is required for various biological processes, and abnormal protein acetylation is associated with various diseases, including NSCLC. Protein deacetylases are currently considered cancer permissive partly due to malignant cells being sensitive to deacetylase inhibition. Sirtuin 2 (SIRT2), a primarily cytosolic nicotinamide adenine dinucleotide-dependent class III protein deacetylase, has been shown to catalyze the removal of acetyl groups from a wide range of proteins, including tubulin, ribonucleotide reductase regulatory subunit M2 and glucose-6-phosphate dehydrogenase. In addition, SIRT2 is also known to possess lysine fatty deacylation activity. Physiologically, SIRT2 serves as a regulator of the cell cycle and of cellular metabolism. It has been shown to play important roles in proliferation, migration and invasion during carcinogenesis. It is notable that both oncogenic and tumor suppressive functions of SIRT2 have been described in NSCLC and other cancer types, suggesting a context-specific role of SIRT2 in cancer progression. In addition, inhibition of SIRT2 exhibits a broad anticancer effect, indicating its potential as a therapeutic for NSCLC tumors with high expression of SIRT2. However, due to the diverse molecular and genetic characteristics of NSCLC, the context-specific function of SIRT2 remains to be determined. The current review investigated the functions of SIRT2 during NSCLC progression with regard to its regulation of metabolism, stem cell-like features and autophagy.
Collapse
Affiliation(s)
- Mengge Zheng
- Institute of Biology and Medical Sciences, Soochow University Medical College, Suzhou, Jiangsu 215123, P.R. China
| | - Changyong Hu
- Institute of Biology and Medical Sciences, Soochow University Medical College, Suzhou, Jiangsu 215123, P.R. China
| | - Meng Wu
- Institute of Biology and Medical Sciences, Soochow University Medical College, Suzhou, Jiangsu 215123, P.R. China
| | - Yue Eugene Chin
- Institute of Biology and Medical Sciences, Soochow University Medical College, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
29
|
Chiu YC, Tseng MC, Hsu CH. Expanding the Substrate Specificity of Macro Domains toward 3″-Isomer of O-Acetyl-ADP-ribose. ACS Catal 2021. [DOI: 10.1021/acscatal.1c01943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yi-Chih Chiu
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan
| | - Mei-Chun Tseng
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Chun-Hua Hsu
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan
- Department of Agricultural Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
30
|
Ghugari R, Tsao S, Schmidt M, Bonneil É, Brenner C, Verreault A. Mechanisms to reduce the cytotoxicity of pharmacological nicotinamide concentrations in the pathogenic fungus Candida albicans. FEBS J 2021; 288:3478-3506. [PMID: 33155404 DOI: 10.1111/febs.15622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/13/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022]
Abstract
Candida albicans is a pathogenic fungus that causes systemic infections and mortality in immunosuppressed individuals. We previously showed that deacetylation of histone H3 lysine 56 by Hst3 is essential for C. albicans viability. Hst3 is a fungal-specific NAD+ -dependent protein deacetylase of the sirtuin family. In vivo, supraphysiological concentrations of nicotinamide (NAM) are required for Hst3 inhibition and cytotoxicity. This underscores the importance of identifying mechanisms by which C. albicans can modulate intracellular NAM concentrations. For the first time in a pathogenic fungus, we combine genetics, heavy isotope labeling, and targeted quantitative metabolomics to identify genes, pathways, and mechanisms by which C. albicans can reduce the cytotoxicity of high NAM concentrations. We discovered three distinct fates for supraphysiological NAM concentrations. First, upon transient exposure to NAM, high intracellular NAM concentrations rapidly return near the physiological levels observed in cells that are not exposed to NAM. Second, during the first step of a fungal-specific NAM salvage pathway, NAM is converted into nicotinic acid, a metabolite that cannot inhibit the sirtuin Hst3. Third, we provide evidence that NAM enters the NAD+ metabolome through a NAM exchange reaction that contributes to NAM-mediated inhibition of sirtuins. However, in contrast to the other fates of NAM, the NAM exchange reaction cannot cause a net decrease in the intracellular concentration of NAM. Therefore, this reaction cannot enhance resistance to NAM. In summary, we demonstrate that C. albicans possesses at least two mechanisms to attenuate the cytotoxicity of pharmacological NAM concentrations. It seems likely that those two mechanisms of resistance to cytotoxic NAM concentrations are conserved in many other pathogenic fungi.
Collapse
Affiliation(s)
- Rahul Ghugari
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
- Programme de Biologie Moléculaire, Université de Montréal, QC, Canada
| | - Sarah Tsao
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
| | - Mark Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, IA, USA
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
| | - Charles Brenner
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Alain Verreault
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, QC, Canada
| |
Collapse
|
31
|
Hou JY, Zhou L, Li JL, Wang DP, Cao JM. Emerging roles of non-histone protein crotonylation in biomedicine. Cell Biosci 2021; 11:101. [PMID: 34059135 PMCID: PMC8166067 DOI: 10.1186/s13578-021-00616-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 05/22/2021] [Indexed: 12/25/2022] Open
Abstract
Crotonylation of proteins is a newly found type of post-translational modifications (PTMs) which occurs leadingly on the lysine residue, namely, lysine crotonylation (Kcr). Kcr is conserved and is regulated by a series of enzymes and co-enzymes including lysine crotonyltransferase (writer), lysine decrotonylase (eraser), certain YEATS proteins (reader), and crotonyl-coenzyme A (donor). Histone Kcr has been substantially studied since 2011, but the Kcr of non-histone proteins is just an emerging field since its finding in 2017. Recent advances in the identification and quantification of non-histone protein Kcr by mass spectrometry have increased our understanding of Kcr. In this review, we summarized the main proteomic characteristics of non-histone protein Kcr and discussed its biological functions, including gene transcription, DNA damage response, enzymes regulation, metabolic pathways, cell cycle, and localization of heterochromatin in cells. We further proposed the performance of non-histone protein Kcr in diseases and the prospect of Kcr manipulators as potential therapeutic candidates in the diseases.
Collapse
Affiliation(s)
- Jia-Yi Hou
- Key Laboratory of Cellular Physiology At Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China.,Department of Clinical Laboratory, Shanxi Provincial Academy of Traditional Chinese Medicine, Taiyuan, China
| | - Lan Zhou
- Key Laboratory of Cellular Physiology At Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jia-Lei Li
- Key Laboratory of Cellular Physiology At Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - De-Ping Wang
- Key Laboratory of Cellular Physiology At Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology At Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
32
|
Spinck M, Bischoff M, Lampe P, Meyer-Almes FJ, Sievers S, Neumann H. Discovery of Dihydro-1,4-Benzoxazine Carboxamides as Potent and Highly Selective Inhibitors of Sirtuin-1. J Med Chem 2021; 64:5838-5849. [PMID: 33876629 DOI: 10.1021/acs.jmedchem.1c00017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sirtuins are signaling hubs orchestrating the cellular response to various stressors with roles in all major civilization diseases. Sirtuins remove acyl groups from lysine residues of proteins, thereby controlling their activity, turnover, and localization. The seven human sirtuins, SirT1-7, are closely related in structure, hindering the development of specific inhibitors. Screening 170,000 compounds, we identify and optimize SirT1-specific benzoxazine inhibitors, Sosbo, which rival the efficiency and surpass the selectivity of selisistat (EX527). The compounds inhibit the deacetylation of p53 in cultured cells, demonstrating their ability to permeate biological membranes. Kinetic analysis of inhibition and docking studies reveal that the inhibitors bind to a complex of SirT1 and nicotinamide adenine dinucleotide, similar to selisistat. These new SirT1 inhibitors are valuable alternatives to selisistat in biochemical and cell biological studies. Their greater selectivity may allow the development of better targeted drugs to combat SirT1 activity in diseases such as cancer, Huntington's chorea, or anorexia.
Collapse
Affiliation(s)
- Martin Spinck
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany
| | - Matthias Bischoff
- Compound Management and Screening Center, Dortmund, Otto-Hahn-Str. 11, Dortmund 44227, Germany
| | - Philipp Lampe
- Compound Management and Screening Center, Dortmund, Otto-Hahn-Str. 11, Dortmund 44227, Germany
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Stephanstrasse 7, Darmstadt 64295, Germany
| | - Sonja Sievers
- Compound Management and Screening Center, Dortmund, Otto-Hahn-Str. 11, Dortmund 44227, Germany
| | - Heinz Neumann
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, Dortmund 44227, Germany.,Department of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Stephanstrasse 7, Darmstadt 64295, Germany
| |
Collapse
|
33
|
Wang M, Lin H. Understanding the Function of Mammalian Sirtuins and Protein Lysine Acylation. Annu Rev Biochem 2021; 90:245-285. [PMID: 33848425 DOI: 10.1146/annurev-biochem-082520-125411] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein lysine acetylation is an important posttranslational modification that regulates numerous biological processes. Targeting lysine acetylation regulatory factors, such as acetyltransferases, deacetylases, and acetyl-lysine recognition domains, has been shown to have potential for treating human diseases, including cancer and neurological diseases. Over the past decade, many other acyl-lysine modifications, such as succinylation, crotonylation, and long-chain fatty acylation, have also been investigated and shown to have interesting biological functions. Here, we provide an overview of the functions of different acyl-lysine modifications in mammals. We focus on lysine acetylation as it is well characterized, and principles learned from acetylation are useful for understanding the functions of other lysine acylations. We pay special attention to the sirtuins, given that the study of sirtuins has provided a great deal of information about the functions of lysine acylation. We emphasize the regulation of sirtuins to illustrate that their regulation enables cells to respond to various signals and stresses.
Collapse
Affiliation(s)
- Miao Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA;
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA; .,Howard Hughes Medical Institute, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
34
|
Cai Q, Tian L, Xie JT, Huang QY, Feng MG, Keyhani NO. A fungal sirtuin modulates development and virulence in the insect pathogen, Beauveria bassiana. Environ Microbiol 2021; 23:5164-5183. [PMID: 33817929 DOI: 10.1111/1462-2920.15497] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 01/02/2023]
Abstract
Chromatin transitions are mediated in part by acetylation/deacetylation post-translational modifications of histones. Histone deacetylases, e.g. sirtuins (Sir-proteins), repress transcription via promotion of heterochromatin formation. Here, we characterize the Sir2 class III histone deacetylase (BbSir2) in the environmentally and economically important fungal insect pathogen, Beauveria bassiana. BbSir2 is shown to contribute to the deacetylation of lysine residues on H3 and H4 histones. Targeted gene knockout of BbSir2 resulted in impaired asexual development, reduced abilities to utilize various carbon/nitrogen sources, reduced tolerance to oxidative, heat, and UV stress, and attenuated virulence. ΔBbSir2 cells showed disrupted cell cycle development and abnormal hyphal septation patterns. Proteomic protein acetylation analyses of wild type and ΔBbSir2 cells revealed the differential abundance of 462 proteins and altered (hyper- or hypo-) acetylation of 436 lysine residues on 350 proteins. Bioinformatic analyses revealed enrichment in pathways involved in carbon/nitrogen metabolism, cell cycle control and cell rescue, defence and mitochondrial functioning. Critical targets involved in virulence included LysM effector proteins and a benzoquinone oxidoreductase implicated in detoxification of cuticular compounds. These data indicate broad effects of BbSir2 on fungal development and stress response, with identification of discrete targets that can account for the observed (decreased) virulence phenotype.
Collapse
Affiliation(s)
- Qing Cai
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.,Department of Microbiology and Cell Science, University of Florida, Bldg. 981, Museum Road, Gainesville, FL, 32611, USA
| | - Li Tian
- Shandong Provincial Key Laboratory of Microbial Engineering, Department of Bioengineering, Qilu University of Technology, Jinan, Shandong, 250353, China
| | - Jia-Tao Xie
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Qiu-Ying Huang
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Ming-Guang Feng
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Nemat O Keyhani
- Department of Microbiology and Cell Science, University of Florida, Bldg. 981, Museum Road, Gainesville, FL, 32611, USA
| |
Collapse
|
35
|
Nicotinamide adenine dinucleotide (NAD+): essential redox metabolite, co-substrate and an anti-cancer and anti-ageing therapeutic target. Biochem Soc Trans 2021; 48:733-744. [PMID: 32573651 DOI: 10.1042/bst20190033] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/10/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) and its reduced form NADH are essential coupled redox metabolites that primarily promote cellular oxidative (catabolic) metabolic reactions. This enables energy generation through glycolysis and mitochondrial respiration to support cell growth and survival. In addition, many key enzymes that regulate diverse cell functions ranging from gene expression to proteostasis require NAD+ as a co-substrate for their catalytic activity. This includes the NAD+-dependent sirtuin family of protein deacetylases and the PARP family of DNA repair enzymes. Whilst their vital activity consumes NAD+ which is cleaved to nicotinamide, several pathways exist for re-generating NAD+ and sustaining NAD+ homeostasis. However, there is growing evidence of perturbed NAD+ homeostasis and NAD+-regulated processes contributing to multiple disease states. NAD+ levels decline in the human brain and other organs with age and this is associated with neurodegeneration and other age-related diseases. Dietary supplementation with NAD+ precursors is being investigated to counteract this. Paradoxically, many cancers have increased dependency on NAD+. Clinical efforts to exploit this have so far shown limited success. Emerging new opportunities to exploit dysregulation of NAD+ metabolism in cancers are critically discussed. An update is also provided on other key NAD+ research including perturbation of the NAD+ salvage enzyme NAMPT in the context of the tumour microenvironment (TME), methodology to study subcellular NAD+ dynamics in real-time and the regulation of differentiation by competing NAD+ pools.
Collapse
|
36
|
Song W, Liu ML, Zhao ZJ, Huang CQ, Xu JW, Wang AQ, Li P, Fan YB. SIRT1 Inhibits High Shear Stress-Induced Apoptosis in Rat Cortical Neurons. Cell Mol Bioeng 2020; 13:621-631. [PMID: 33281991 PMCID: PMC7704980 DOI: 10.1007/s12195-020-00623-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/03/2020] [Indexed: 10/24/2022] Open
Abstract
INTRODUCTION Sirtuin1 (SIRT1), one of NAD+-dependent protein deacetylases, is proved to be neuroprotective in aging diseases, but its effect on neuronal apoptosis has not been clarified. To investigate the role of SIRT1 in inhibiting neuronal apoptosis, SIRT1 was interfered or overexpressed in cortical neurons. METHODS We exerted overloading laminar shear stress with 10 dyn/cm2 for 4, 8, and 12 h on neurons to cause cortical neuronal apoptosis, and the apoptosis percentage was tested by TUNEL assay. The adenovirus plasmids containing SIRT1 RNA interference or SIRT1 wild type gene were transfected into neurons before shear stress loading. SIRT1 mRNA and protein level were tested by Real-time PCR, immunofluorescence and western blots assay. RESULTS SIRT1 was primarily expressed in nucleus of cortical neurons, and its mRNA level was significantly increased after 4 h stimulation. SIRT1 RNAi cortical neurons had higher TUNEL positive cells, while SIRT1 overexpression significantly decreased the percentage of died cells induced by shear stress compared to control group. CONCLUSIONS SIRT1 plays a neuroprotective role in shear stress induced apoptosis and could be as potential pharmacological targets against neuronal degeneration in future.
Collapse
Affiliation(s)
- Wei Song
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - Mei-Li Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - Zhi-Jun Zhao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - Chong-Quan Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - Jun-Wei Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - An-Qing Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - Ping Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
| | - Yu-Bo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191 China
- National Research Center for Rehabilitation Technical Aids, Beijing, 100176 China
| |
Collapse
|
37
|
Teixeira CSS, Cerqueira NMFSA, Gomes P, Sousa SF. A Molecular Perspective on Sirtuin Activity. Int J Mol Sci 2020; 21:ijms21228609. [PMID: 33203121 PMCID: PMC7696986 DOI: 10.3390/ijms21228609] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
The protein acetylation of either the α-amino groups of amino-terminal residues or of internal lysine or cysteine residues is one of the major posttranslational protein modifications that occur in the cell with repercussions at the protein as well as at the metabolome level. The lysine acetylation status is determined by the opposing activities of lysine acetyltransferases (KATs) and lysine deacetylases (KDACs), which add and remove acetyl groups from proteins, respectively. A special group of KDACs, named sirtuins, that require NAD+ as a substrate have received particular attention in recent years. They play critical roles in metabolism, and their abnormal activity has been implicated in several diseases. Conversely, the modulation of their activity has been associated with protection from age-related cardiovascular and metabolic diseases and with increased longevity. The benefits of either activating or inhibiting these enzymes have turned sirtuins into attractive therapeutic targets, and considerable effort has been directed toward developing specific sirtuin modulators. This review summarizes the protein acylation/deacylation processes with a special focus on the current developments in the sirtuin research field.
Collapse
Affiliation(s)
- Carla S. S. Teixeira
- UCIBIO/REQUIMTE, BioSIM - Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.S.T.); (N.M.F.S.A.C.)
| | - Nuno M. F. S. A. Cerqueira
- UCIBIO/REQUIMTE, BioSIM - Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.S.T.); (N.M.F.S.A.C.)
| | - Pedro Gomes
- Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal;
- Center for Health Technology and Services Research (CINTESIS), University of Porto, R. Dr. Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Sérgio F. Sousa
- UCIBIO/REQUIMTE, BioSIM - Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.S.T.); (N.M.F.S.A.C.)
- Correspondence: ; Tel.: +351-22-551-3600
| |
Collapse
|
38
|
Ferreira LM, Li AM, Serafim TL, Sobral MC, Alpoim MC, Urbano AM. Intermediary metabolism: An intricate network at the crossroads of cell fate and function. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165887. [DOI: 10.1016/j.bbadis.2020.165887] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/01/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
|
39
|
Shao LW, Peng Q, Dong M, Gao K, Li Y, Li Y, Li CY, Liu Y. Histone deacetylase HDA-1 modulates mitochondrial stress response and longevity. Nat Commun 2020; 11:4639. [PMID: 32934238 PMCID: PMC7493924 DOI: 10.1038/s41467-020-18501-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/24/2020] [Indexed: 01/06/2023] Open
Abstract
The ability to detect, respond and adapt to mitochondrial stress ensures the development and survival of organisms. Caenorhabditis elegans responds to mitochondrial stress by activating the mitochondrial unfolded protein response (UPRmt) to buffer the mitochondrial folding environment, rewire the metabolic state, and promote innate immunity and lifespan extension. Here we show that HDA-1, the C. elegans ortholog of mammalian histone deacetylase (HDAC) is required for mitochondrial stress-mediated activation of UPRmt. HDA-1 interacts and coordinates with the genome organizer DVE-1 to induce the transcription of a broad spectrum of UPRmt, innate immune response and metabolic reprogramming genes. In rhesus monkey and human tissues, HDAC1/2 transcript levels correlate with the expression of UPRmt genes. Knocking down or pharmacological inhibition of HDAC1/2 disrupts the activation of the UPRmt and the mitochondrial network in mammalian cells. Our results underscore an evolutionarily conserved mechanism of HDAC1/2 in modulating mitochondrial homeostasis and regulating longevity.
Collapse
Affiliation(s)
- Li-Wa Shao
- Beijing Advanced Innovation Center for Genomics, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Qi Peng
- Laboratory of Bioinformatics and Genomic Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China
| | - Mingyue Dong
- Beijing Advanced Innovation Center for Genomics, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Kaiyu Gao
- Beijing Advanced Innovation Center for Genomics, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China
| | - Yumei Li
- Laboratory of Bioinformatics and Genomic Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China
| | - Yi Li
- Beijing Advanced Innovation Center for Genomics, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Chuan-Yun Li
- Laboratory of Bioinformatics and Genomic Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China.
| | - Ying Liu
- Beijing Advanced Innovation Center for Genomics, State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, 100871, Beijing, China.
| |
Collapse
|
40
|
Zhang Z, Chen X, Liu S. Role of Sirtuin-1 in Neonatal Hypoxic-Ischemic Encephalopathy and Its Underlying Mechanism. Med Sci Monit 2020; 26:e924544. [PMID: 32826847 PMCID: PMC7461657 DOI: 10.12659/msm.924544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/21/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic encephalopathy (HIE) is a dreaded disease and one of the leading causes of severe neurological dysfunction in neonates. The present study explored the functions of Sirtuin-1 (SIRT1) in neonatal HIE. MATERIAL AND METHODS A HIE neonatal rat model was generated to determine SIRT1 levels in brain tissues. Cell apoptosis and cell viability were analyzed by flow cytometry and MTT assay. qRT-PCR and Western blot analysis were used to assess gene mRNA and protein levels. Subsequently, the effect of SIRT1 on HIE was investigated in vitro by constructing an oxygen-glucose deprivation (OGD) cell model. RESULTS The effective construction of the HIE rat model was confirmed by the enhanced brain cell apoptosis and the increased expression of HIE-related molecular markers, including S100 calcium-binding protein B (S100B) and neuron-specific enolase (NSE). SIRT1 expression was downregulated in HIE rat brain tissues. These findings indicated that SIRT1 was downregulated in neuronal cells subjected to OGD. In addition, enhanced cell viability and reduced cell apoptosis were observed, suggesting that SIRT1 overexpression relieved OGD-induced neuronal cell injury. Transfection with SIRT1-siRNA further increased OGD-induced neuronal cell injury, evidenced by decreased cell viability and enhanced cell apoptosis. Finally, SIRT1 overexpression significantly downregulated p-p65 protein expression. CONCLUSIONS Our findings revealed that SIRT1 may be a novel and promising therapy target for HIE treatment.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R, China
| | - Xin Chen
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, P.R, China
| | - Sichen Liu
- Department of Neonatology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, P.R. China
| |
Collapse
|
41
|
Jiang SB, Lu YS, Liu T, Li LM, Wang HX, Wu Y, Gao XH, Chen HD. UVA influenced the SIRT1-miR-27a-5p-SMAD2-MMP1/COL1/BCL2 axis in human skin primary fibroblasts. J Cell Mol Med 2020; 24:10027-10041. [PMID: 32790210 PMCID: PMC7520305 DOI: 10.1111/jcmm.15610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Both SIRT1 and UVA radiation are involved in cellular damage processes such as apoptosis, senescence and ageing. MicroRNAs (miRNAs) have been reported to be closely related to UV radiation, as well as to SIRT1. In this study, we investigated the connections among SIRT1, UVA and miRNA in human skin primary fibroblasts. Our results showed that UVA altered the protein level of SIRT1 in a time point–dependent manner. Using miRNA microarray, bioinformatics analysis, we found that knocking down SIRT1 could cause up‐regulation of miR‐27a‐5p and the latter could down‐regulate SMAD2, and these results were verified by qRT‐PCR or Western blot. Furthermore, UVA radiation (5 J/cm2), knocking down SIRT1 or overexpression of miR‐27a‐5p led to increased expression of MMP1, and decreased expressions of COL1 and BCL2. We also found additive impacts on MMP1, COL1 and BCL2 under the combination of UVA radiation + Sirtinol (SIRT1 inhibitor), or UVA radiation + miR‐27a‐5p mimic. SIRT1 activator resveratrol could reverse damage changes caused by UVA radiation. Besides, absent of SIRT1 or overexpression of miR‐27a‐5p increased cell apoptosis and induced cell arrest in G2/M phase. Taken together, these results demonstrated that UVA could influence a novel SIRT1‐miR‐27a‐5p‐SMAD2‐MMP1/COL1/BCL2 axis in skin primary fibroblasts, and may provide potential therapeutic targets for UVA‐induced skin damage.
Collapse
Affiliation(s)
- Shi-Bin Jiang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Yan-Song Lu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Tao Liu
- Department of Urinary Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Liang-Man Li
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - He-Xiao Wang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Yan Wu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Xing-Hua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Hong-Duo Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
42
|
Kulikova VA, Nikiforov AA. Role of NUDIX Hydrolases in NAD and ADP-Ribose Metabolism in Mammals. BIOCHEMISTRY. BIOKHIMIIA 2020; 85:883-894. [PMID: 33045949 DOI: 10.1134/s0006297920080040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
Proteins of the NUDIX hydrolase (NUDT) superfamily that cleave organic pyrophosphates are found in all classes of organisms, from archaea and bacteria to higher eukaryotes. In mammals, NUDTs exhibit a wide range of functions and are characterized by different substrate specificity and intracellular localization. They control the concentration of various metabolites in the cell, including key regulatory molecules such as nicotinamide adenine dinucleotide (NAD), ADP-ribose, and their derivatives. In this review, we discuss the role of NUDT proteins in the metabolism of NAD and ADP-ribose in human and animal cells.
Collapse
Affiliation(s)
- V A Kulikova
- Peter the Great St. Petersburg Polytechnic University, Saint Petersburg, 195251, Russia.
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, 194064, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, 194223, Russia
| | - A A Nikiforov
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, 194064, Russia
| |
Collapse
|
43
|
Spinck M, Neumann‐Staubitz P, Ecke M, Gasper R, Neumann H. Evolvierte, selektive Eraser für spezifische Lysinacylierungen. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Martin Spinck
- Abteilung StrukturbiochemieMax-Planck-Institut für molekulare Physiologie Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Petra Neumann‐Staubitz
- Abteilung StrukturbiochemieMax-Planck-Institut für molekulare Physiologie Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Maria Ecke
- Abteilung StrukturbiochemieMax-Planck-Institut für molekulare Physiologie Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Raphael Gasper
- Abteilung Kristallographie und BiophysikMax-Planck-Institut für molekulare Physiologie Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Heinz Neumann
- Abteilung StrukturbiochemieMax-Planck-Institut für molekulare Physiologie Otto-Hahn-Straße 11 44227 Dortmund Deutschland
- Fachbereich Chemie- und BiotechnologieTechnische Hochschule Darmstadt Stephanstraße 7 64295 Darmstadt Deutschland
| |
Collapse
|
44
|
Spinck M, Neumann‐Staubitz P, Ecke M, Gasper R, Neumann H. Evolved, Selective Erasers of Distinct Lysine Acylations. Angew Chem Int Ed Engl 2020; 59:11142-11149. [PMID: 32187803 PMCID: PMC7317389 DOI: 10.1002/anie.202002899] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/16/2020] [Indexed: 12/31/2022]
Abstract
Lysine acylations, a family of diverse protein modifications varying in acyl-group length, charge, and saturation, are linked to many important physiological processes. Only a small set of substrate-promiscuous lysine acetyltransferases and deacetylases (KDACs) install and remove this vast variety of modifications. Engineered KDACs that remove only one type of acylation would help to dissect the different contributions of distinct acylations. We developed a bacterial selection system for the directed evolution of KDACs and identified variants up to 400 times more selective for butyryl-lysine compared to crotonyl-lysine. Structural analyses revealed that the enzyme adopts different conformational states depending on the type of acylation of the bound peptide. We used the butyryl-selective KDAC variant to shift the cellular acylation spectrum towards increased lysine crotonylation. These new enzymes will help in dissecting the roles of different lysine acylations in cell physiology.
Collapse
Affiliation(s)
- Martin Spinck
- Department of Structural BiochemistryMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Petra Neumann‐Staubitz
- Department of Structural BiochemistryMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Maria Ecke
- Department of Structural BiochemistryMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Raphael Gasper
- Crystallography and Biophysics UnitMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Heinz Neumann
- Department of Structural BiochemistryMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstrasse 764295DarmstadtGermany
| |
Collapse
|
45
|
Moraes DS, Moreira DC, Andrade JMO, Santos SHS. Sirtuins, brain and cognition: A review of resveratrol effects. IBRO Rep 2020; 9:46-51. [PMID: 33336103 PMCID: PMC7733131 DOI: 10.1016/j.ibror.2020.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/20/2020] [Indexed: 02/08/2023] Open
Abstract
Sirtuins (SIRTs) are a protein family with high preservation degree among evolutionary scale. SIRTs are histone deacetylases regulatory enzymes of genetic material deeply involved in numerous physiological tasks including metabolism, brain function and aging. Mammals sirtuins comprise seven enzymatic components (SIRT1–SIRT7). The highest studied sirtuin is SIRT1, which plays an essential position in the prevention and evolution of neuro-disorders. Resveratrol (3,5,4-trihydroxystylbene) (RSV) is a polyphenol, which belongs to a family compounds identified as stilbenes, predominantly concentrated in grapes and red wine. RSV is the must studied Sirtuin activator and is used as food supplementary compound. Resveratrol exhibits strong antioxidant activity, reducing free radicals, diminishing quinone-reductase-2 activity and exerting positive regulation of several endogenous enzymes. Resveratrol is also able to inhibit pro-inflammatory factors, reducing the stimulation of the nuclear factor kB (NF-kB) and the release of endogenous cytokines. Resveratrol treatment can modulate multiple signaling pathway effectors related to programmed cell death, cell survival, and synaptic plasticity. In this context, the present review looks over news and the role of Sirtuins activation and resveratrol effects on modulating target genes, cognition and neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel Silva Moraes
- Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Daniele Cristina Moreira
- Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - João Marcus Oliveira Andrade
- Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil.,Institute of Agricultural Sciences (ICA), Food Engineering, Universidade Federal de Minas Gerais, Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
46
|
Klein MA, Denu JM. Biological and catalytic functions of sirtuin 6 as targets for small-molecule modulators. J Biol Chem 2020; 295:11021-11041. [PMID: 32518153 DOI: 10.1074/jbc.rev120.011438] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Sirtuin 6 (SIRT6) is a nuclear NAD+-dependent deacetylase of histone H3 that regulates genome stability and gene expression. However, nonhistone substrates and additional catalytic activities of SIRT6, including long-chain deacylation and mono-ADP-ribosylation of other proteins, have also been reported, but many of these noncanonical roles remain enigmatic. Genetic studies have revealed critical homeostatic cellular functions of SIRT6, underscoring the need to better understand which catalytic functions and molecular pathways are driving SIRT6-associated phenotypes. At the physiological level, SIRT6 activity promotes increased longevity by regulating metabolism and DNA repair. Recent work has identified natural products and synthetic small molecules capable of activating the inefficient in vitro deacetylase activity of SIRT6. Here, we discuss the cellular functions of SIRT6 with a focus on attributing its catalytic activity to its proposed biological functions. We cover the molecular architecture and catalytic mechanisms that distinguish SIRT6 from other NAD+-dependent deacylases. We propose that combining specific SIRT6 amino acid substitutions identified in enzymology studies and activity-selective compounds could help delineate SIRT6 functions in specific biological contexts and resolve the apparently conflicting roles of SIRT6 in processes such as tumor development. We further highlight the recent development of small-molecule modulators that provide additional biological insight into SIRT6 functions and offer therapeutic approaches to manage metabolic and age-associated diseases.
Collapse
Affiliation(s)
- Mark A Klein
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin, USA.,Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - John M Denu
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin, USA .,Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
47
|
Chandrasekaran K, Choi J, Arvas MI, Salimian M, Singh S, Xu S, Gullapalli RP, Kristian T, Russell JW. Nicotinamide Mononucleotide Administration Prevents Experimental Diabetes-Induced Cognitive Impairment and Loss of Hippocampal Neurons. Int J Mol Sci 2020; 21:ijms21113756. [PMID: 32466541 PMCID: PMC7313029 DOI: 10.3390/ijms21113756] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetes predisposes to cognitive decline leading to dementia and is associated with decreased brain NAD+ levels. This has triggered an intense interest in boosting nicotinamide adenine dinucleotide (NAD+) levels to prevent dementia. We tested if the administration of the precursor of NAD+, nicotinamide mononucleotide (NMN), can prevent diabetes-induced memory deficits. Diabetes was induced in Sprague-Dawley rats by the administration of streptozotocin (STZ). After 3 months of diabetes, hippocampal NAD+ levels were decreased (p = 0.011). In vivo localized high-resolution proton magnetic resonance spectroscopy (MRS) of the hippocampus showed an increase in the levels of glucose (p < 0.001), glutamate (p < 0.001), gamma aminobutyric acid (p = 0.018), myo-inositol (p = 0.018), and taurine (p < 0.001) and decreased levels of N-acetyl aspartate (p = 0.002) and glutathione (p < 0.001). There was a significant decrease in hippocampal CA1 neuronal volume (p < 0.001) and neuronal number (p < 0.001) in the Diabetic rats. Diabetic rats showed hippocampal related memory deficits. Intraperitoneal NMN (100 mg/kg) was given after induction and confirmation of diabetes and was provided on alternate days for 3 months. NMN increased brain NAD+ levels, normalized the levels of glutamate, taurine, N-acetyl aspartate (NAA), and glutathione. NMN-treatment prevented the loss of CA1 neurons and rescued the memory deficits despite having no significant effect on hyperglycemic or lipidemic control. In hippocampal protein extracts from Diabetic rats, SIRT1 and PGC-1α protein levels were decreased, and acetylation of proteins increased. NMN treatment prevented the diabetes-induced decrease in both SIRT1 and PGC-1α and promoted deacetylation of proteins. Our results indicate that NMN increased brain NAD+, activated the SIRT1 pathway, preserved mitochondrial oxidative phosphorylation (OXPHOS) function, prevented neuronal loss, and preserved cognition in Diabetic rats.
Collapse
Affiliation(s)
- Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Joungil Choi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA;
| | - Muhammed Ikbal Arvas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Mohammad Salimian
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Sujal Singh
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.X.); (R.P.G.)
| | - Rao P Gullapalli
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.X.); (R.P.G.)
| | - Tibor Kristian
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA;
- Department of Anesthesiology; University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James William Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA;
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence:
| |
Collapse
|
48
|
Impact of Nutrition on Short-Term Exercise-Induced Sirtuin Regulation: Vegans Differ from Omnivores and Lacto-Ovo Vegetarians. Nutrients 2020; 12:nu12041004. [PMID: 32260570 PMCID: PMC7230533 DOI: 10.3390/nu12041004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
Keywords: sirtuins; vegetarian; vegan; exercise; endurance athletes; metabolic regulation.
Collapse
|
49
|
Hypothalamic NAD +-Sirtuin Axis: Function and Regulation. Biomolecules 2020; 10:biom10030396. [PMID: 32143417 PMCID: PMC7175325 DOI: 10.3390/biom10030396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
The rapidly expanding elderly population and obesity endemic have become part of continuing global health care problems. The hypothalamus is a critical center for the homeostatic regulation of energy and glucose metabolism, circadian rhythm, and aging-related physiology. Nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase sirtuins are referred to as master metabolic regulators that link the cellular energy status to adaptive transcriptional responses. Mounting evidence now indicates that hypothalamic sirtuins are essential for adequate hypothalamic neuronal functions. Owing to the NAD+-dependence of sirtuin activity, adequate hypothalamic NAD+ contents are pivotal for maintaining energy homeostasis and circadian physiology. Here, we comprehensively review the regulatory roles of the hypothalamic neuronal NAD+-sirtuin axis in a normal physiological context and their changes in obesity and the aging process. We also discuss the therapeutic potential of NAD+ biology-targeting drugs in aging/obesity-related metabolic and circadian disorders.
Collapse
|
50
|
Feng X, Wang Y, Chen W, Xu S, Li L, Geng Y, Shen A, Gao H, Zhang L, Liu S. SIRT3 inhibits cardiac hypertrophy by regulating PARP-1 activity. Aging (Albany NY) 2020; 12:4178-4192. [PMID: 32139662 PMCID: PMC7093179 DOI: 10.18632/aging.102862] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/24/2020] [Indexed: 01/14/2023]
Abstract
Sirtuin 3 (SIRT3) is a type III histone deacetylase that inhibits cardiac hypertrophy. It is mainly localized in the mitochondria and is thus implicated in mitochondrial metabolism. Recent studies have shown that SIRT3 can also accumulate in the nuclear under stressed conditions, and participated in histone deacetylation of target proteins. Poly [ADP-ribose] polymerase 1 (PARP-1) functions as an important PARP isoform that was involved in cardiac hypertrophy. Our experiments showed that SIRT3 accumulated in the nuclear of cardiomyocytes treated with isoproterenol or SIRT3 overexpression. Moreover, overexpression of SIRT3 by adenovirus inhibited the expression of cardiac hypertrophic genes-ANF and BNP, as well as abrogating PARP-1 activation induced by isoproterenol or phenylephrine. In addition, co-immunoprecipitation experiments revealed that SIRT3 could interact with PARP-1, and overexpression of SIRT3 could decrease the acetylation level of PARP-1. Our results indicate that SIRT3 exerts protective effects against cardiac hypertrophy by reducing the level of acetylation and activity of PARP-1, thus providing novel mechanistic insights into SIRT3-mediated cardiprotective actions.
Collapse
Affiliation(s)
- Xiaojun Feng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Yanan Wang
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, PR. China
| | - Wenxu Chen
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, PR. China
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, West Henrietta, NY 14586, USA
| | - Lingli Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Yadi Geng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Aizong Shen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China.,Anhui Provincial Cardiovascular Institute, Hefei, Anhui, PR. China
| | - Hui Gao
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, PR. China
| | - Lei Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Sheng Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| |
Collapse
|