1
|
Modulation of SIRT6 activity acts as an emerging therapeutic implication for pathological disorders in the skeletal system. Genes Dis 2022. [DOI: 10.1016/j.gendis.2021.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
2
|
Gupta A, Hwang BJ, Benyamien-Roufaeil D, Jain S, Liu S, Gonzales R, Brown RA, Zalzman M, Lu AL, Lu AL, University of Maryland School of Medicine, Baltimore, MD, USA, University of Maryland School of Medicine, Baltimore, MD, USA, University of Maryland School of Medicine, Baltimore, MD, USA, University of Maryland School of Medicine, Baltimore, MD, USA, University of Maryland School of Medicine, Baltimore, MD, USA, University of Maryland School of Medicine, Baltimore, MD, USA, University of Maryland School of Medicine; The Center for Stem Cell Biology and Regenerative Medicine; Marlene and Stewart Greenbaum Cancer Center, Baltimore, MD 21201, USA, University of Maryland School of Medicine; Marlene and Stewart Greenbaum Cancer Center, Baltimore, MD, USA. Mammalian MutY Homolog (MYH or MUTYH) is Critical for Telomere Integrity under Oxidative Stress. OBM GERIATRICS 2022; 6:196. [PMID: 35812693 PMCID: PMC9267527 DOI: 10.21926/obm.geriatr.2202196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Telomeres consist of special features and proteins to protect the ends of each chromosome from deterioration and fusion. The telomeric DNA repeats are highly susceptible to oxidative damage that can accelerate telomere shortening and affect telomere integrity. Several DNA repair factors including MYH/MUTYH DNA glycosylase, its interacting partners Rad9/Rad1/Hus1 checkpoint clamp, and SIRT6 aging regulator, are associated with the telomeres. MYH prevents C:G to A:T mutation by removing adenine mispaired with a frequent oxidative DNA lesion, 8-oxoguanine. Here, we show that hMYH knockout (KO) human HEK-293T cells are more sensitive to H2O2 treatment, have higher levels of DNA strand breaks and shorter telomeres than the control hMYH +/+ cells. SIRT6 foci increase at both the global genome and at telomeric regions in H2O2-treated hMYH +/+ cells. However, in untreated hMYH KO HEK-293T cells, SIRT6 foci only increase at the global genome, but not at the telomeric regions. In addition, the hMYH KO HEK-293T cells have increased extra-chromosomal and intra-chromosomal telomeres compared to the control cells, even in the absence of H2O2 treatment. After H2O2 treatment, the frequency of extra-chromosomal telomeres increased in control HEK-293T cells. Remarkably, in H2O2-treated hMYH KO cells, the frequencies of extra-chromosomal telomeres, intra-chromosomal telomeres, and telomere fusions are further increased. We further found that the sensitivity to H2O2 and shortened telomeres of hMYH KO cells, are restored by expressing wild-type hMYH, and partially rescued by expressing hMYHQ324H mutant (defective in Hus1 interaction only), but not by expressing hMYHV315A mutant (defective in both SIRT6 and Hus1 interactions). Thus, MYH interactions with SIRT6 and Hus1 are critical for maintaining cell viability and telomeric stability. Therefore, the failure to coordinate 8-oxoG repair is detrimental to telomere integrity.
Collapse
Affiliation(s)
- Aditi Gupta
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bor-Jang Hwang
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Sara Jain
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sophie Liu
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rex Gonzales
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert A Brown
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michal Zalzman
- University of Maryland School of Medicine; The Center for Stem Cell Biology and Regenerative Medicine; Marlene and Stewart Greenbaum Cancer Center, Baltimore, MD 21201, USA
| | - A-Lien Lu
- University of Maryland School of Medicine; Marlene and Stewart Greenbaum Cancer Center, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Gorkhali R, Tian L, Dong B, Bagchi P, Deng X, Pawar S, Duong D, Fang N, Seyfried N, Yang J. Extracellular calcium alters calcium-sensing receptor network integrating intracellular calcium-signaling and related key pathway. Sci Rep 2021; 11:20576. [PMID: 34663830 PMCID: PMC8523568 DOI: 10.1038/s41598-021-00067-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are a target for over 34% of current drugs. The calcium-sensing receptor (CaSR), a family C GPCR, regulates systemic calcium (Ca2+) homeostasis that is critical for many physiological, calciotropical, and noncalciotropical outcomes in multiple organs. However, the mechanisms by which extracellular Ca2+ (Ca2+ex) and the CaSR mediate networks of intracellular Ca2+-signaling and players involved throughout the life cycle of CaSR are largely unknown. Here we report the first CaSR protein–protein interactome with 94 novel putative and 8 previously published interactors using proteomics. Ca2+ex promotes enrichment of 66% of the identified CaSR interactors, pertaining to Ca2+ dynamics, endocytosis, degradation, trafficking, and primarily to protein processing in the endoplasmic reticulum (ER). These enhanced ER-related processes are governed by Ca2+ex-activated CaSR which directly modulates ER-Ca2+ (Ca2+ER), as monitored by a novel ER targeted Ca2+-sensor. Moreover, we validated the Ca2+ex dependent colocalizations and interactions of CaSR with ER-protein processing chaperone, 78-kDa glucose regulated protein (GRP78), and with trafficking-related protein. Live cell imaging results indicated that CaSR and vesicle-associated membrane protein-associated A (VAPA) are inter-dependent during Ca2+ex induced enhancement of near-cell membrane expression. This study significantly extends the repertoire of the CaSR interactome and reveals likely novel players and pathways of CaSR participating in Ca2+ER dynamics, agonist mediated ER-protein processing and surface expression.
Collapse
Affiliation(s)
- Rakshya Gorkhali
- Department of Chemistry, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Li Tian
- Department of Chemistry, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Bin Dong
- Department of Chemistry, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Xiaonan Deng
- Department of Chemistry, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Shrikant Pawar
- Department of Biology, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Duc Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ning Fang
- Department of Chemistry, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Nicholas Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jenny Yang
- Department of Chemistry, Center of Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
4
|
Shi L, Wang Y, Oppong TB, Fu X, Yang H, Wang Y. Prognostic role of SIRT6 in gastrointestinal cancers: a meta-analysis. Open Med (Wars) 2020; 15:358-365. [PMID: 33335996 PMCID: PMC7712292 DOI: 10.1515/med-2020-0403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/15/2022] Open
Abstract
Sirtuin 6 (SIRT6) plays a critical role in the progression and development of gastrointestinal cancers. However, the association between SIRT6 expression and clinicopathological parameters and prognosis in gastrointestinal cancer patients remains inconclusive. Consequently, we conducted this meta-analysis to evaluate the importance of SIRT6 expression in various types of gastrointestinal cancers. PubMed, EMBASE, and Web of Science databases were systematically searched to screen the relevant literature. The reported or estimated hazard ratio (HR) and odds ratio (OR) and their corresponding 95% confidence interval (CI) were pooled to assess the strength of the association. Nine studies involving 867 patients were included in the meta-analysis. Overall analysis showed that high SIRT6 expression was related to better overall survival in gastrointestinal cancers (HR = 0.62, 95% CI = 0.47–0.82). High SIRT6 expression was also related to a favorable tumor node metastasis (TNM) stage (OR = 0.44, 95% CI = 0.28–0.70) among gastrointestinal cancer patients. Our meta-analysis revealed that high SIRT6 expression might be a potential biomarker predicting better prognosis in gastrointestinal cancers, which may offer options for gastrointestinal cancer treatment.
Collapse
Affiliation(s)
- Li Shi
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Ying Wang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Timothy Bonney Oppong
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaoli Fu
- Department of Social Medicine and Health Management, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| |
Collapse
|
5
|
An ordered assembly of MYH glycosylase, SIRT6 protein deacetylase, and Rad9-Rad1-Hus1 checkpoint clamp at oxidatively damaged telomeres. Aging (Albany NY) 2020; 12:17761-17785. [PMID: 32991318 PMCID: PMC7585086 DOI: 10.18632/aging.103934] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/07/2020] [Indexed: 01/24/2023]
Abstract
In the base excision repair pathway, MYH/MUTYH DNA glycosylase prevents mutations by removing adenine mispaired with 8-oxoG, a frequent oxidative lesion. MYH glycosylase activity is enhanced by Rad9-Rad1-Hus1 (9-1-1) checkpoint clamp and SIRT6 histone/protein deacetylase. Here, we show that MYH, SIRT6, and 9-1-1 are recruited to confined oxidatively damaged regions on telomeres in mammalian cells. Using different knockout cells, we show that SIRT6 responds to damaged telomeres very early, and then recruits MYH and Hus1 following oxidative stress. However, the recruitment of Hus1 to damaged telomeres is partially dependent on SIRT6. The catalytic activities of SIRT6 are not important for SIRT6 response but are essential for MYH recruitment to damaged telomeres. Compared to wild-type MYH, the recruitment of hMYHV315A mutant (defective in both SIRT6 and Hus1 interactions), but not hMYHQ324H mutant (defective in Hus1 interaction only), to damaged telomeres is severely reduced. The formation of MYH/SIRT6/9-1-1 complex is of biological significance as interrupting their interactions can increase cell's sensitivity to H2O2 and/or elevate cellular 8-oxoG levels after H2O2 treatment. Our results establish that SIRT6 acts as an early sensor of BER enzymes and both SIRT6 and 9-1-1 serve critical roles in DNA repair to maintain telomere integrity.
Collapse
|
6
|
Al-Azzam N. Sirtuin 6 and metabolic genes interplay in Warburg effect in cancers. J Clin Biochem Nutr 2020; 66:169-175. [PMID: 32523242 DOI: 10.3164/jcbn.19-110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/13/2019] [Indexed: 01/10/2023] Open
Abstract
Under oxygen availability, normal cells undergo mitochondrial oxidative phosphorylation to metabolize glucose and yield up to 36 ATPs per glucose molecule for cellular functions, and undergo non-oxidative metabolism (glycolysis) under hypoxic and proliferating conditions to yield 2 ATP per glucose. These cells metabolize glucose to pyruvate via glycolysis followed by conversion of pyruvate to lactate via lactate dehydrogenase. However, cancer cells have the ability to undergo glycolysis and ferment glucose to lactate regardless of oxygen availability; a phenomenon first addressed by Otto Warburg and called, "Warburg effect". Numerous glycolytic genes/proteins have been identified in tumors; that include glucose transporter 1 (GLUT1), hexokinase 2 (HK2), pyruvate kinase-M2 splice isoform (PKM2), and lactate dehydrogenase (LDH-A). Histone deacetylase sirtuin 6 (SIRT6), an epigenetic regulator, is highly expressed in various cancers. SIRT6 plays an important role in Warburg effect by regulating many glycolytic genes. Loss of SIRT6 enhances tumor growth via enhancing glycolysis. This review is mainly concerned with exploring the most recent advances in understanding the roles of the metabolic genes (GLUT1, HK2, PKM2, and LDH-A) and the epigenetic regulator SIRT6 in cancer metabolism and how SIRT6 can modulate these metabolic genes expression and its possible use as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Nosayba Al-Azzam
- Department of Physiology and Biochemistry, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| |
Collapse
|
7
|
The molecular mechanisms associated with PIN7, a protein-protein interaction network of seven pleiotropic proteins. J Theor Biol 2020; 487:110124. [DOI: 10.1016/j.jtbi.2019.110124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 01/12/2023]
|
8
|
Chang AR, Ferrer CM, Mostoslavsky R. SIRT6, a Mammalian Deacylase with Multitasking Abilities. Physiol Rev 2020; 100:145-169. [PMID: 31437090 PMCID: PMC7002868 DOI: 10.1152/physrev.00030.2018] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 08/14/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Mammalian sirtuins have emerged in recent years as critical modulators of multiple biological processes, regulating cellular metabolism, DNA repair, gene expression, and mitochondrial biology. As such, they evolved to play key roles in organismal homeostasis, and defects in these proteins have been linked to a plethora of diseases, including cancer, neurodegeneration, and aging. In this review, we describe the multiple roles of SIRT6, a chromatin deacylase with unique and important functions in maintaining cellular homeostasis. We attempt to provide a framework for such different functions, for the ability of SIRT6 to interconnect chromatin dynamics with metabolism and DNA repair, and the open questions the field will face in the future, particularly in the context of putative therapeutic opportunities.
Collapse
Affiliation(s)
- Andrew R Chang
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts; and The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Christina M Ferrer
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts; and The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts; and The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
9
|
Current role of mammalian sirtuins in DNA repair. DNA Repair (Amst) 2019; 80:85-92. [DOI: 10.1016/j.dnarep.2019.06.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/29/2019] [Indexed: 01/20/2023]
|
10
|
Abstract
Cell reprogramming has been considered a powerful technique in the regenerative medicine field. In addition to diverse its strengths, cell reprogramming technology also has several drawbacks generated during the process of reprogramming. Telomere shortening caused by the cell reprogramming process impedes the efficiency of cell reprogramming. Transcription factors used for reprogramming alter genomic contents and result in genetic mutations. Additionally, defective mitochondria functioning such as excessive mitochondrial fission leads to the limitation of pluripotency and ultimately reduces the efficiency of reprogramming. These problems including genomic instability and impaired mitochondrial dynamics should be resolved to apply cell reprograming in clinical research and to address efficiency and safety concerns. Sirtuin (NAD+-dependent histone deacetylase) has been known to control the chromatin state of the telomere and influence mitochondria function in cells. Recently, several studies reported that Sirtuins could control for genomic instability in cell reprogramming. Here, we review recent findings regarding the role of Sirtuins in cell reprogramming. And we propose that the manipulation of Sirtuins may improve defects that result from the steps of cell reprogramming.
Collapse
Affiliation(s)
- Jaein Shin
- Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University, Seoul 04620, Korea
| | - Junyeop Kim
- Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University, Seoul 04620, Korea
| | - Hanseul Park
- Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University, Seoul 04620, Korea
| | - Jongpil Kim
- Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University, Seoul 04620, and Department of Chemistry, Dongguk University, Seoul 04620, Korea
| |
Collapse
|
11
|
Avrahami EM, Levi S, Zajfman E, Regev C, Ben-David O, Arbely E. Reconstitution of Mammalian Enzymatic Deacylation Reactions in Live Bacteria Using Native Acylated Substrates. ACS Synth Biol 2018; 7:2348-2354. [PMID: 30207693 PMCID: PMC6198279 DOI: 10.1021/acssynbio.8b00314] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Lysine deacetylases
(KDACs) are enzymes that catalyze the hydrolysis
of acyl groups from acyl-lysine residues. The recent identification
of thousands of putative acylation sites, including specific acetylation
sites, created an urgent need for biochemical methodologies aimed
at better characterizing KDAC-substrate specificity and evaluating
KDACs activity. To address this need, we utilized genetic code expansion
technology to coexpress site-specifically acylated substrates with
mammalian KDACs, and study substrate recognition and deacylase activity
in live Escherichia coli. In this system the bacterial
cell serves as a “biological test tube” in which the
incubation of a single mammalian KDAC and a potential peptide or full-length
acylated substrate transpires. We report novel deacetylation activities
of Zn2+-dependent deacetylases and sirtuins in bacteria.
We also measure the deacylation of propionyl-, butyryl-, and crotonyl-lysine,
as well as novel deacetylation of Lys310-acetylated RelA by SIRT3,
SIRT5, SIRT6, and HDAC8. This study highlights the importance of native
interactions to KDAC-substrate recognition and deacylase activity.
Collapse
Affiliation(s)
- Emanuel M. Avrahami
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shahar Levi
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Eyal Zajfman
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Clil Regev
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Oshrit Ben-David
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Eyal Arbely
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
12
|
Bai L, Lin G, Sun L, Liu Y, Huang X, Cao C, Guo Y, Xie C. Upregulation of SIRT6 predicts poor prognosis and promotes metastasis of non-small cell lung cancer via the ERK1/2/MMP9 pathway. Oncotarget 2018; 7:40377-40386. [PMID: 27777384 PMCID: PMC5130014 DOI: 10.18632/oncotarget.9750] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 05/20/2016] [Indexed: 12/16/2022] Open
Abstract
Sirtuin6 (SIRT6), a member of the sirtuins protein family, plays multiple complex roles in cancer. Here, we report that elevated SIRT6 expression was correlated with clinicopathological parameters such as T and N classification in non-small cell lung cancer (NSCLC) patient tumors. SIRT6 overexpression in NSCLC cell lines increased extracellular signal-regulated kinase (p-ERK)1/2 phosphorylation, activated matrix metalloproteinase 9 (MMP9) and promoted tumor cell migration and invasion. Upon treatment with a specific mitogen-activated protein kinase (MEK) 1/2 inhibitor, these effects were abolished. Our results demonstrate SIRT6 upregulation in NSCLC for the first time and suggest a functional role for SIRT6 in promoting migration and invasion through ERK1/2/MMP9 signaling. SIRT6 may serve as a potential therapeutic target in NSCLC and its utility as a prognostic indicator warrants further study.
Collapse
Affiliation(s)
- Lihong Bai
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Gengpeng Lin
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Longhua Sun
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.,Respiratory Department, Nanchang Hospital of Integrative Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Yangli Liu
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xinyan Huang
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chuangjie Cao
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yubiao Guo
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Canmao Xie
- Respiratory Department, The First Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
13
|
Lin H, Hao Y, Zhao Z, Tong Y. Sirtuin 6 contributes to migration and invasion of osteosarcoma cells via the ERK1/2/MMP9 pathway. FEBS Open Bio 2017; 7:1291-1301. [PMID: 28904859 PMCID: PMC5586348 DOI: 10.1002/2211-5463.12265] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/08/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022] Open
Abstract
Dysregulation of sirtuin 6 (SIRT6) is actively involved in tumor progression. High levels of SIRT6 have been associated with hepatocellular carcinoma and non‐small cell lung cancer, and SIRT6 facilitates growth and metastasis of cancer cells. However, the clinical significance and biological function of SIRT6 are not known for osteosarcoma (OS). Here, we report that SIRT6 was notably overexpressed in OS tissues compared with non‐cancerous specimens. The high level of SIRT6 was prominently correlated with malignant clinical parameters and poor prognosis of OS patients. SIRT6 was also up‐regulated in OS cells. SIRT6 knockdown inhibited the invasion and migration of Saos‐2 and U2OS cells in vitro, while SIRT6 restoration increased these cellular biological behaviors in MG‐63 cells. Mechanistically, SIRT6 up‐regulated expression of matrix metallopeptidase 9 (MMP9) in OS cells. MMP9 restoration partially abolished the effects of SIRT6 knockdown on OS cells, with increased cell migration and invasion. MMP9 knockdown reduced migration and invasion of SIRT6‐overexpressing MG‐63 cells. Furthermore, SIRT6 positively modulated the levels of phosphorylated extracellular signal‐regulated kinases 1 and 2 (ERK1/2). PD098059 and PD0325901, inhibitors of mitogen‐activated protein kinase kinase (MEK), blocked the regulatory effects of SIRT6 on p‐ERK1/2 and MMP9 levels, suggesting that SIRT6 regulated MMP9 abundance probably through the MEK–ERK1/2 pathway. These results suggest that SIRT6 may act as a prognostic predictor and a drug target for OS patients.
Collapse
Affiliation(s)
- Hang Lin
- Department of Orthopedics Zhejiang Hospital Hangzhou China
| | - Yi Hao
- Department of Orthopedics Zhejiang Hospital Hangzhou China
| | - Zhengxu Zhao
- Department of Orthopedics Zhejiang Hospital Hangzhou China
| | - Yongjun Tong
- Department of Orthopedics Zhejiang Hospital Hangzhou China
| |
Collapse
|
14
|
Nahálková J. The protein-interaction network with functional roles in tumorigenesis, neurodegeneration, and aging. Mol Cell Biochem 2016; 423:187-196. [DOI: 10.1007/s11010-016-2836-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/23/2016] [Indexed: 01/13/2023]
|
15
|
Hwang BJ, Jin J, Gao Y, Shi G, Madabushi A, Yan A, Guan X, Zalzman M, Nakajima S, Lan L, Lu AL. SIRT6 protein deacetylase interacts with MYH DNA glycosylase, APE1 endonuclease, and Rad9-Rad1-Hus1 checkpoint clamp. BMC Mol Biol 2015; 16:12. [PMID: 26063178 PMCID: PMC4464616 DOI: 10.1186/s12867-015-0041-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/29/2015] [Indexed: 02/07/2023] Open
Abstract
Background SIRT6, a member of the NAD+-dependent histone/protein deacetylase family, regulates genomic stability, metabolism, and lifespan. MYH glycosylase and APE1 are two base excision repair (BER) enzymes involved in mutation avoidance from oxidative DNA damage. Rad9–Rad1–Hus1 (9–1–1) checkpoint clamp promotes cell cycle checkpoint signaling and DNA repair. BER is coordinated with the checkpoint machinery and requires chromatin remodeling for efficient repair. SIRT6 is involved in DNA double-strand break repair and has been implicated in BER. Here we investigate the direct physical and functional interactions between SIRT6 and BER enzymes. Results We show that SIRT6 interacts with and stimulates MYH glycosylase and APE1. In addition, SIRT6 interacts with the 9-1-1 checkpoint clamp. These interactions are enhanced following oxidative stress. The interdomain connector of MYH is important for interactions with SIRT6, APE1, and 9–1–1. Mutagenesis studies indicate that SIRT6, APE1, and Hus1 bind overlapping but different sequence motifs on MYH. However, there is no competition of APE1, Hus1, or SIRT6 binding to MYH. Rather, one MYH partner enhances the association of the other two partners to MYH. Moreover, APE1 and Hus1 act together to stabilize the MYH/SIRT6 complex. Within human cells, MYH and SIRT6 are efficiently recruited to confined oxidative DNA damage sites within transcriptionally active chromatin, but not within repressive chromatin. In addition, Myh foci induced by oxidative stress and Sirt6 depletion are frequently localized on mouse telomeres. Conclusions Although SIRT6, APE1, and 9-1-1 bind to the interdomain connector of MYH, they do not compete for MYH association. Our findings indicate that SIRT6 forms a complex with MYH, APE1, and 9-1-1 to maintain genomic and telomeric integrity in mammalian cells. Electronic supplementary material The online version of this article (doi:10.1186/s12867-015-0041-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bor-Jang Hwang
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA.
| | - Jin Jin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA.
| | - Ying Gao
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA. .,School of Medicine, Tsinghua University, No.1 Tsinghua Yuan, Haidian District, Beijing, 100084, China.
| | - Guoli Shi
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA. .,University of Maryland School of Nursing, 655 West Lombard Street, Baltimore, MD, 21201, USA.
| | - Amrita Madabushi
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA. .,Department of Natural and Physical Sciences, Life Sciences Institute, Baltimore City Community College, 801 West Baltimore Street, Baltimore, MD, 21201, USA.
| | - Austin Yan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA.
| | - Xin Guan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA.
| | - Michal Zalzman
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA. .,Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, 16 South Eutaw Street, Baltimore, MD, 21201, USA.
| | - Satoshi Nakajima
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA. .,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Li Lan
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA. .,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - A-Lien Lu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA. .,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
16
|
Jenwitheesuk A, Nopparat C, Mukda S, Wongchitrat P, Govitrapong P. Melatonin regulates aging and neurodegeneration through energy metabolism, epigenetics, autophagy and circadian rhythm pathways. Int J Mol Sci 2014; 15:16848-84. [PMID: 25247581 PMCID: PMC4200827 DOI: 10.3390/ijms150916848] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/03/2014] [Accepted: 09/12/2014] [Indexed: 12/19/2022] Open
Abstract
Brain aging is linked to certain types of neurodegenerative diseases and identifying new therapeutic targets has become critical. Melatonin, a pineal hormone, associates with molecules and signaling pathways that sense and influence energy metabolism, autophagy, and circadian rhythms, including insulin-like growth factor 1 (IGF-1), Forkhead box O (FoxOs), sirtuins and mammalian target of rapamycin (mTOR) signaling pathways. This review summarizes the current understanding of how melatonin, together with molecular, cellular and systemic energy metabolisms, regulates epigenetic processes in the neurons. This information will lead to a greater understanding of molecular epigenetic aging of the brain and anti-aging mechanisms to increase lifespan under healthy conditions.
Collapse
Affiliation(s)
- Anorut Jenwitheesuk
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| | - Chutikorn Nopparat
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| | - Prapimpun Wongchitrat
- Center for Innovation Development and Technology Transfer, Faculty of Medical Technology, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| |
Collapse
|
17
|
Lee N, Kim DK, Kim ES, Park SJ, Kwon JH, Shin J, Park SM, Moon YH, Wang HJ, Gho YS, Choi KY. Comparative interactomes of SIRT6 and SIRT7: Implication of functional links to aging. Proteomics 2014; 14:1610-22. [PMID: 24782448 DOI: 10.1002/pmic.201400001] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/01/2014] [Accepted: 04/25/2014] [Indexed: 12/21/2022]
Abstract
Sirtuins are NAD(+) -dependent deacetylases that regulate a range of cellular processes. Although diverse functions of sirtuins have been proposed, those functions of SIRT6 and SIRT7 that are mediated by their interacting proteins remain elusive. In the present study, we identified SIRT6- and SIRT7-interacting proteins, and compared their interactomes to investigate functional links. Our interactomes revealed 136 interacting proteins for SIRT6 and 233 for SIRT7 while confirming seven and 111 proteins identified previously for SIRT6 and SIRT7, respectively. Comparison of SIRT6 and SIRT7 interactomes under the same experimental conditions disclosed 111 shared proteins, implying related functional links. The interaction networks of interactomes indicated biological processes associated with DNA repair, chromatin assembly, and aging. Interactions of two highly acetylated proteins, nucleophosmin (NPM1) and nucleolin, with SIRT6 and SIRT7 were confirmed by co-immunoprecipitation. NPM1 was found to be deacetylated by both SIRT6 and SIRT7. In senescent cells, the acetylation level of NPM1 was increased in conjunction with decreased levels of SIRT6 and SIRT7, suggesting that the acetylation of NPM1 could be regulated by SIRT6 and SIRT7 in the aging process. Our comparative interactomic study of SIRT6 and SIRT7 implies important functional links to aging by their associations with interacting proteins. All MS data have been deposited in the ProteomeXchange with identifiers PXD000159 and PXD000850 (http://proteomecentral.proteomexchange.org/dataset/PXD000159, http://proteomecentral.proteomexchange.org/dataset/PXD000850).
Collapse
Affiliation(s)
- Namgyu Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Favero G, Rezzani R, Rodella LF. Sirtuin 6 nuclear localization at cortical brain level of young diabetic mice: an immunohistochemical study. Acta Histochem 2014; 116:272-7. [PMID: 24063863 DOI: 10.1016/j.acthis.2013.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/05/2013] [Accepted: 08/06/2013] [Indexed: 01/15/2023]
Abstract
A study was conducted to assess the physiopathological significance of sirtuin 6 (SIRT6) at the brain cortical level. We analyzed the specific expression and subcellular localization of SIRT6 in young db/db mice, an experimental animal model of type II Diabetes mellitus. We analyzed the cytoarchitecture of the brain cortex, evaluated SIRT6 expression and its localization by immunohistochemistry comparing db/db mice to lean control mice, examining the six cortical layers and the motor and somatosensory cortex. Finally, we calculated a SIRT6 labeling index. We observed the absence of significant morphological differences between lean and db/db mice, indicating that young db/db mice showed a neuronal morphology and distribution similar to that of lean mice and also normal brain tissue architecture with intact cortical layers. Moreover, sirtuin 6 is mainly localized in the nucleus of both lean and db/db mice. In particular, the db/db mice showed few positive cells compared to lean control mice in all cortical layers. We found a lower sirtuin 6 labeling index without significant differences between the motor and somatosensory cortex. Our findings contribute to further understanding the sirtuin 6 immunohistochemical changes in the early stages of type II Diabetes mellitus and propose its possible implication in the pathogenic processes associated with Diabetes mellitus and diabetes-induced neurodegeneration.
Collapse
|
19
|
Miteva YV, Cristea IM. A proteomic perspective of Sirtuin 6 (SIRT6) phosphorylation and interactions and their dependence on its catalytic activity. Mol Cell Proteomics 2013; 13:168-83. [PMID: 24163442 DOI: 10.1074/mcp.m113.032847] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Sirtuin 6 (SIRT6), a member of the mammalian sirtuin family, is a nuclear deacetylase with substrate-specific NAD(+)-dependent activity. SIRT6 has emerged as a critical regulator of diverse processes, including DNA repair, gene expression, telomere maintenance, and metabolism. However, our knowledge regarding its interactions and regulation remains limited. Here, we present a comprehensive proteomics-based analysis of SIRT6 protein interactions and their dependence on SIRT6 catalytic activity. We also identify evolutionarily conserved SIRT6 phosphorylations, including four within a proline-rich disordered region, and show that the conserved S338 phosphorylation can modulate selected SIRT6 interactions. By integrating molecular biology tools, microscopy, immunoaffinity purifications, label-free quantitative mass spectrometry, and bioinformatic analyses, we have established the first large-scale SIRT6 interaction network. Relative protein abundances and gene ontology functional assessment highlighted proteins involved in transcription regulation, chromatin organization, nuclear transport, telomerase function, and RNA processing. Independent immunoisolations under increased stringency distinguished the most stable SIRT6 interactions. One prominent interaction with Ras-GTPase-activating protein-binding protein 1 (G3BP1) was further validated by microscopy, reciprocal purifications, and isolations in different cell types and of endogenous SIRT6. Interestingly, a subset of specific interactions, including G3BP1, were significantly reduced or abolished in isolations of catalytically deficient SIRT6 mutant, revealing previously unknown interplay between SIRT6 activity and its associations. Overall, our study reveals putative means of regulation of SIRT6 functions via interactions and modifications, providing an important resource for future studies on the molecular mechanisms underlying sirtuin functions.
Collapse
Affiliation(s)
- Yana V Miteva
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | | |
Collapse
|
20
|
Chai Z, Dai A, Tu Y, Li J, Wu T, Wang Y, Hale LJ, Koentgen F, Thomas MC, Cooper ME. Genetic deletion of cell division autoantigen 1 retards diabetes-associated renal injury. J Am Soc Nephrol 2013; 24:1782-92. [PMID: 23929772 DOI: 10.1681/asn.2013010060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Cell division autoantigen 1 (CDA1) enhances TGF-β signaling in renal and vascular cells, and renal expression of CDA1 is elevated in animal models of diabetes. In this study, we investigated the genetic deletion of Tspyl2, the gene encoding CDA1, in C57BL6 and ApoE knockout mice. The increased renal expression of TGF-β1, TGF-β type I and II receptors, and phosphorylated Smad3 associated with diabetes in wild-type mice was attenuated in diabetic CDA1 knockout mice. Notably, CDA1 deletion significantly reduced diabetes-associated renal matrix accumulation and immunohistochemical staining for collagens III and IV and attenuated glomerular and tubulointerstitial injury indices, despite the presence of persistent hyperglycemia, polyuria, renal hypertrophy, and hyperfiltration. Furthermore, CDA1 deletion reduced gene expression of TGF-β1 receptors in the kidney, resulting in a functionally attenuated response to exogenous TGF-β, including reduced levels of phosphorylated Smad3 and ERK1/2, in primary kidney cells from CDA1 knockout animals. Taken together, these data suggest that CDA1 deletion reduces but does not block renal TGF-β signaling. Because direct antagonism of TGF-β or its receptors has unwanted effects, CDA1 may be a potential therapeutic target for retarding DN and perhaps, other kidney diseases associated with TGF-β-mediated fibrogenesis.
Collapse
|
21
|
The role of SIRT1 in ocular aging. Exp Eye Res 2013; 116:17-26. [PMID: 23892278 DOI: 10.1016/j.exer.2013.07.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 07/13/2013] [Accepted: 07/16/2013] [Indexed: 12/27/2022]
Abstract
The sirtuins are a highly conserved family of nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases that helps regulate the lifespan of diverse organisms. The human genome encodes seven different sirtuins (SIRT1-7), which share a common catalytic core domain but possess distinct N- and C-terminal extensions. Dysfunction of some sirtuins have been associated with age-related diseases, such as cancer, type II diabetes, obesity-associated metabolic diseases, neurodegeneration, and cardiac aging, as well as the response to environmental stress. SIRT1 is one of the targets of resveratrol, a polyphenolic SIRT1 activator that has been shown to increase the lifespan and to protect various organs against aging. A number of animal studies have been conducted to examine the role of sirtuins in ocular aging. Here we review current knowledge about SIRT1 and ocular aging. The available data indicate that SIRT1 is localized in the nucleus and cytoplasm of cells forming all normal ocular structures, including the cornea, lens, iris, ciliary body, and retina. Upregulation of SIRT1 has been shown to have an important protective effect against various ocular diseases, such as cataract, retinal degeneration, optic neuritis, and uveitis, in animal models. These results suggest that SIRT1 may provide protection against diseases related to oxidative stress-induced ocular damage, including cataract, age-related macular degeneration, and optic nerve degeneration in glaucoma patients.
Collapse
|