1
|
Chen Y, Lu S, Shan S, Wu W, He X, Farag MA, Chen W, Zhao C. New insights into phytochemicals via protein glycosylation focused on aging and diabetes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156673. [PMID: 40220419 DOI: 10.1016/j.phymed.2025.156673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/29/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Protein glycosylation as a common post-translational modification that has significant impacts on protein folding, enzymatic activity, and interfering with receptor functioning. In recent years, with the rapid development of glycopeptide enrichment and analysis technology and the deepening of glycosylation research, glycosylation has gradually become a sign of disease occurrence and development. Multiple investigations suggest that protein glycosylation affect the advances of diabetes and aging. PURPOSE AND METHODS This review was focused on the action mechanisms of glycosylated proteins production, permanent abnormalities in extracellular matrix component function, inflammatory and reactive oxygen species production, as well as the glycosylated characterizations of diabetes and aging. Further, advances in glycosylation analysis and detection methods are presented for the first time, highlighting for needed future developments. All literatures were gathered from PubMed and Google Scholar. RESULTS Herein, we review how protein glycosylation impacts the progression of diabetes and aging. Specifically, we focus on various types of glycosylation, including N-linked glycosylation, O-linked glycosylation, C-glycosylation, S-glycosylation, and glycophosphatidylinositol (GPI) anchors. N-linked glycosylation and O-linked glycosylation are commonly observed glycosylation forms, wherein O-GlcNAcylation plays a significant role in diabetes, while N-glycan could serve as biomarkers for identifying inflammation and aging. CONCLUSIONS Protein glycosylation produces a vastly larger number of core glycan structures through utilizing at least 173 glycosyltransferases and repeated common scaffolds. Single protein may contain multiple glycosylation sites, and the structure and occupancy of glycan at each site may be different, resulting in the macro heterogeneity of protein glycosylation. This review will contribute to how protein glycosylation impacts the life progress of cells and its association with diseases.
Collapse
Affiliation(s)
- Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Suyue Lu
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shuo Shan
- University of Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E32004 Ourense, Spain
| | - Weihao Wu
- College of Food Engineering, Zhangzhou Institute of Technology, Zhangzhou 363000, China
| | - Xinxin He
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Weichao Chen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
2
|
Yu W, Hu S, Xu X, Shao J, Yan J, Ding F. Association between O-GlcNAc transferase activity and major adverse cardiovascular events: findings from the China PEACE-MPP cohort. BMC Cardiovasc Disord 2025; 25:281. [PMID: 40221667 PMCID: PMC11992785 DOI: 10.1186/s12872-025-04732-6] [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: 04/12/2024] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The O-GlcNAc transferase (OGT) levels are closely related to the O-GlcNAcylation of proteins and are also associated with cardiovascular disease. This study explored the association between OGT activity and major adverse cardiovascular events (MACEs) in patients with high cardiovascular disease risk. This post hoc study included patients from the China PEACE-MPP study in Yi Wu, Zhejiang Province, between 2014 and 2015. METHODS The patients were divided into the low and high OGT activity groups according to the median serum OGT value. The outcome was the occurrence of MACEs (cardiovascular death, non-fatal acute myocardial infarction, and non-fatal ischemic stroke). RESULTS Finally, 1947 participants (973 and 974 with low and high OGT activity, respectively) were included. The mean follow-up was 5.56 ± 1.01 years. The participants in the low OGT activity group had a significantly higher occurrence rate of MACEs compared with the high OGT activity group (100 [10.4%] vs. 74 [7.6%], P = 0.032). The Kaplan-Meier analysis showed that the event-free survival rate in the low OGT activity group was significantly lower than in the high OGT activity group (P = 0.036). Multivariable Cox proportional hazards regression analysis showed that after adjustment for age, drinking, hyperglycemia, history of hypertension, and history of cardiovascular and cerebrovascular disease, a high OGT activity was independently associated with a lower risk of MACEs (HR = 0.738, 95%CI: 0.547-0.997, P = 0.048). CONCLUSIONS A low OGT activity was independently associated with an increased risk of MACE among patients with a high risk of cardiovascular disease. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Wei Yu
- Zhejiang Provincial Center for Cardiovascular Disease Control and Prevention, Zhejiang Hospital, Hangzhou, 310013, China
| | - Shiyun Hu
- Zhejiang Provincial Center for Cardiovascular Disease Control and Prevention, Zhejiang Hospital, Hangzhou, 310013, China
| | - Xiaoling Xu
- Zhejiang Provincial Center for Cardiovascular Disease Control and Prevention, Zhejiang Hospital, Hangzhou, 310013, China
| | - Jianlin Shao
- Zhejiang Provincial Center for Cardiovascular Disease Control and Prevention, Zhejiang Hospital, Hangzhou, 310013, China
| | - Jing Yan
- Zhejiang Provincial Center for Cardiovascular Disease Control and Prevention, Zhejiang Hospital, Hangzhou, 310013, China
| | - Fang Ding
- Zhejiang Provincial Center for Cardiovascular Disease Control and Prevention, Zhejiang Hospital, Hangzhou, 310013, China.
| |
Collapse
|
3
|
Lin C, Xia M, Dai Y, Huang Q, Sun Z, Zhang G, Luo R, Peng Q, Li J, Wang X, Lin H, Gao X, Tang H, Shen X, Wang S, Jin L, Hao X, Zheng Y. Cross-ancestry analyses of Chinese and European populations reveal insights into the genetic architecture and disease implication of metabolites. CELL GENOMICS 2025; 5:100810. [PMID: 40118068 PMCID: PMC12008806 DOI: 10.1016/j.xgen.2025.100810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025]
Abstract
Differential susceptibilities to various diseases and corresponding metabolite variations have been documented across diverse ethnic populations, but the genetic determinants of these disparities remain unclear. Here, we performed large-scale genome-wide association studies of 171 directly quantifiable metabolites from a nuclear magnetic resonance-based metabolomics platform in 10,792 Han Chinese individuals. We identified 15 variant-metabolite associations, eight of which were successfully replicated in an independent Chinese population (n = 4,480). By cross-ancestry meta-analysis integrating 213,397 European individuals from the UK Biobank, we identified 228 additional variant-metabolite associations and improved fine-mapping precision. Moreover, two-sample Mendelian randomization analyses revealed evidence that genetically predicted levels of triglycerides in high-density lipoprotein were associated with a higher risk of coronary artery disease and that of glycine with a lower risk of heart failure in both ancestries. These findings enhance our understanding of the shared and specific genetic architecture of metabolites as well as their roles in complex diseases across populations.
Collapse
Affiliation(s)
- Chenhao Lin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Guoqing Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; National Genomics Data Center& Bio-Med Big Data Center, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai 200031, China
| | - Ruijin Luo
- Shanghai Southgene Technology Co., Ltd., Shanghai 201203, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinxi Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xiaofeng Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan University-the People's Hospital of Rugao Joint Research Institute of Longevity and Aging, Rugao, Jiangsu 226500, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xia Shen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, Guangdong 511400, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Bolanle IO, Palmer TM. O-GlcNAcylation and Phosphorylation Crosstalk in Vascular Smooth Muscle Cells: Cellular and Therapeutic Significance in Cardiac and Vascular Pathologies. Int J Mol Sci 2025; 26:3303. [PMID: 40244145 PMCID: PMC11989994 DOI: 10.3390/ijms26073303] [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: 01/27/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
More than 400 different types of post-translational modifications (PTMs), including O-GlcNAcylation and phosphorylation, combine to co-ordinate almost all aspects of protein function. Often, these PTMs overlap and the specific relationship between O-GlcNAcylation and phosphorylation has drawn much attention. In the last decade, the significance of this dynamic crosstalk has been linked to several chronic pathologies of cardiovascular origin. However, very little is known about the pathophysiological significance of this crosstalk for vascular smooth muscle cell dysfunction in cardiovascular disease. O-GlcNAcylation occurs on serine and threonine residues which are also targets for phosphorylation. A growing body of research has now emerged linking altered vascular integrity and homeostasis with highly regulated crosstalk between these PTMs. Additionally, a significant body of evidence indicates that O-GlcNAcylation is an important contributor to the pathogenesis of neointimal hyperplasia and vascular restenosis responsible for long-term vein graft failure. In this review, we evaluate the significance of this dynamic crosstalk and its role in cardiovascular pathologies, and the prospects of identifying possible targets for more effective therapeutic interventions.
Collapse
Affiliation(s)
| | - Timothy M. Palmer
- Biomedical Institute for Multimorbidity, Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| |
Collapse
|
5
|
Jiang Y, Cai W, Lei G, Cai G, Wu Q, Lu P. Deubiquitinase USP47 Ameliorates Cardiac Hypertrophy Through Reducing Protein O-GlcNAcylation. J Cardiovasc Pharmacol 2025; 85:54-62. [PMID: 39436323 DOI: 10.1097/fjc.0000000000001640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/07/2024] [Indexed: 10/23/2024]
Abstract
ABSTRACT Cardiac hypertrophy is a crucial risk factor for heart failure when the heart is confronted with physiologic or pathologic stimuli. The ubiquitin-proteasome system plays a critical role in the pathogenesis of cardiac hypertrophy. However, as a key component of the ubiquitin-proteasome system, the role of deubiquitinating enzymes in cardiac hypertrophy is not well understood. In this study, we observed that the expression level of deubiquitinase USP47 was increased in hypertrophic hearts and angiotensin II (Ang II)-stimulated neonatal rat cardiomyocytes. Adenovirus-mediated gain- and loss-of-function approaches indicated that USP47 overexpression significantly attenuated Ang II-induced cardiac hypertrophy in vitro and in vivo, whereas endogenous USP47 deficiency promoted the prohypertrophic effect of Ang II. Further investigation demonstrated that USP47 inhibited O-GlcNAcylation in cardiomyocytes by controlling the expression of O-GlcNAcase. Mechanistically, USP47 bound, deubiquitinated, and stabilized protein arginine methyltransferase 5 (PRMT5), thus upregulating O-GlcNAcase expression. We found that the restoration of PRMT5 abolished the prohypertrophic effects of USP47 silence in vitro. Therefore, our results provide the first evidence of the involvement of USP47 in cardiac hypertrophy and identify USP47 as a potential target for hypertrophic therapy.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China ; and
| | - Wenyao Cai
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China ; and
| | - Guangtao Lei
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Guorong Cai
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China ; and
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Peng Lu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
Sreedevi K, James A, Do S, Yedla S, Arowa S, Oka SI, Wende AR, Zaitsev AV, Warren JS. PERM1 regulates mitochondrial energetics through O-GlcNAcylation in the heart. J Mol Cell Cardiol 2025; 198:1-12. [PMID: 39581161 DOI: 10.1016/j.yjmcc.2024.11.002] [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: 05/10/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
PERM1 was initially identified as a new downstream target of PGC-1α and ERRs that regulates mitochondrial bioenergetics in skeletal muscle. Subsequently, we and other groups demonstrated that PERM1 is also a positive regulator of mitochondrial bioenergetics in the heart. However, the exact mechanisms of regulatory functions of PERM1 remain poorly understood. O-GlcNAcylation is a post-translational modification of proteins that are regulated by two enzymes: O-GlcNAc transferase (OGT) that adds O-GlcNAc to proteins; O-GlcNAcase (OGA) that removes O-GlcNAc from proteins. O-GlcNAcylation is a powerful signaling mechanism mediating cellular responses to stressors and nutrient availability, which, among other targets, may influence cardiac metabolism. We hypothesized that PERM1 regulates mitochondrial energetics in cardiomyocytes through modulation of O-GlcNAcylation. We found that overexpression of PERM1 decreased the total levels of O-GlcNAcylated proteins, concomitant with decreased OGT and increased OGA expression levels. Luciferase gene reporter assay showed that PERM1 significantly decreases the promoter activity of Ogt without changing the promoter activity of Oga. The downregulation of OGT by PERM1 overexpression was mediated through its interaction with E2F1, a known transcription repressor of Ogt. A deliberate increase of O-GlcNAcylation through Oga silencing in cardiomyocytes decreased the basal and maximal mitochondrial respiration and ATP production rates, all of which were completely restored by PERM1 overexpression. Furthermore, excessive O-GlcNAcylation caused by the loss of PERM1 led to the increase of O-GlcNAcylated PGC-1α, a master regulator of mitochondrial bioenergetics, concurrent with the dissociation of PGC-1α from PPARα, a well-known transcription factor that regulates fatty acid β-oxidation. We conclude that PERM1 positively regulates mitochondrial energetics, in part, via suppressing O-GlcNAcylation in cardiac myocytes.
Collapse
Affiliation(s)
- Karthi Sreedevi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Amina James
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Sara Do
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Shreya Yedla
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Sumaita Arowa
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Shin-Ichi Oka
- Departiment of Cell and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexey V Zaitsev
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Junco S Warren
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA; Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, USA; Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Na HJ, Kim JM, Kim Y, Lee SH, Sung MJ. Magnolia kobus DC. Regulates Vascular Smooth Muscle Cell Proliferation by Modulating O-GlcNAc and MOF Expression. Prev Nutr Food Sci 2024; 29:430-440. [PMID: 39759825 PMCID: PMC11699568 DOI: 10.3746/pnf.2024.29.4.430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) undergo metabolic pathway transitions, including aerobic glycolysis, fatty acid oxidation, and amino acid metabolism, which are important for their function. Metabolic dysfunction in VSMCs can lead to age-related vascular diseases. O-GlcNAcylation, a nutrient-dependent posttranslational modification linked specifically to glucose metabolism, plays an important role in this context. Magnolia kobus DC. (MK), derived from the flower buds of Magnolia biondii, is known for its anticancer, anti-allergy, and anti-inflammatory properties. However, the role of O-GlcNAcylation in VSMCs under aging and the association between MK and O-GlcNAc remain unclear. Therefore, the present study aimed to determine the effects of O-GlcNAc on VSMC proliferation, along with the expression of MOF (males absent on the first, KAT8) and its correlation with the efficacy of MK. The results showed that aging and O-GlcNAc induction increased the expression levels of O-GlcNAc, O-GlcNAc transferase (OGT), ataxia telangiectasia mutated (ATM) protein, and MOF in mouse vascular smooth muscle cells (MOVAS) and aorta tissue. Transfection with OGT siRNA reduced the expression of MOF and OGT, indicating that OGT regulates MOF and influences cell proliferation. MK treatment reduced the expression of OGT, ATM, and MOF, which was correlated with O-GlcNAc levels. These findings suggest that O-GlcNAcylation is important for VSMC homeostasis and may be a novel target for vascular diseases. Thus, MK exhibits potential as a new drug candidate for treating vascular diseases by modulating O-GlcNAcylation and MOF interactions.
Collapse
Affiliation(s)
- Hyun-Jin Na
- Aging and Metabolism Research Group, Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea
| | - Jong Min Kim
- Aging and Metabolism Research Group, Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea
| | - Yiseul Kim
- Aging and Metabolism Research Group, Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea
| | - Sang Hee Lee
- Aging and Metabolism Research Group, Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea
| | - Mi-Jeong Sung
- Aging and Metabolism Research Group, Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea
| |
Collapse
|
8
|
Zhang YY, Zhang SY, Hu ZX, Voglmeir J, Liu L, Galan MC, Ghirardello M. High sensitivity profiling of N-glycans from mouse serum using fluorescent imidazolium tags by HILIC electrospray ionisation spectrometry. Carbohydr Polym 2024; 343:122449. [PMID: 39174089 DOI: 10.1016/j.carbpol.2024.122449] [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: 03/27/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 08/24/2024]
Abstract
N-linked glycosylation is a ubiquitous protein post-translational modification in which aberrant glycan biosynthesis has been linked to severe conditions like cancer. Accurate qualitative and quantitative analysis of N-glycans are crucial for investigating their physiological functions. Owing to the intrinsic absence of chromophores and high polarity of the glycans, current detection methods are restricted to liquid chromatography and mass spectrometry. Herein, we describe three new imidazolium-based glycan tags: 2'GITag, 3'GITag, and 4'GITag, that significantly improve both the limit of detection and limit of quantification of derivatized oligosaccharides, in terms of fluorescence intensity and ionisation efficiency. Our top-performing derivatisation agent, 4'GITag, shifted the detection sensitivity range from high femtomole to sub-femtomole levels in ESI-MS compared to traditional glycan label, 2AB, enabling the identification of 24 N-glycans in mouse serum, including those bearing sialic acids. Additionally, 4'GITag stabilized Na-salt forms of sialic acids, simplifying the simultaneous analysis of neutral and negative charged N-glycans significantly, avoiding the need for complex derivatisation procedures typically required for the detection of sialylated species. Overall, the favorable performance of imidazolium tags in the derivatisation and sensitive profiling of glycans has the potential for labeling tissue or live cells to explore disease biomarkers and for developing new targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yao-Yao Zhang
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China; Lipid Technology and Engineering, School of Food Science and Engineering, Henan University of Technology, Lianhua Road 100, 450001 Zhengzhou, China; School of Chemistry, University of Bristol, Cantock's Close, BS8 1TS Bristol, UK
| | - Si-Yu Zhang
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - Zi-Xuan Hu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - Josef Voglmeir
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China.
| | - M Carmen Galan
- School of Chemistry, University of Bristol, Cantock's Close, BS8 1TS Bristol, UK.
| | - Mattia Ghirardello
- School of Chemistry, University of Bristol, Cantock's Close, BS8 1TS Bristol, UK; Department of Chemistry, Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Universidad de La Rioja, 26006 Logroño, La Rioja, Spain.
| |
Collapse
|
9
|
Zhu Q, Li J, Sun H, Fan Z, Hu J, Chai S, Lin B, Wu L, Qin W, Wang Y, Hsieh-Wilson LC, Yi W. O-GlcNAcylation of enolase 1 serves as a dual regulator of aerobic glycolysis and immune evasion in colorectal cancer. Proc Natl Acad Sci U S A 2024; 121:e2408354121. [PMID: 39446384 PMCID: PMC11536113 DOI: 10.1073/pnas.2408354121] [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: 04/29/2024] [Accepted: 09/26/2024] [Indexed: 10/27/2024] Open
Abstract
Aerobic glycolysis and immune evasion are two key hallmarks of cancer. However, how these two features are mechanistically linked to promote tumor growth is not well understood. Here, we show that the glycolytic enzyme enolase-1 (ENO1) is dynamically modified with an O-linked β-N-acetylglucosamine (O-GlcNAcylation), and simultaneously regulates aerobic glycolysis and immune evasion via differential glycosylation. Glycosylation of threonine 19 (T19) on ENO1 promotes its glycolytic activity via the formation of active dimers. On the other hand, glycosylation of serine 249 (S249) on ENO1 inhibits its interaction with PD-L1, decreases association of PD-L1 with the E3 ligase STUB1, resulting in stabilization of PD-L1. Consequently, blockade of T19 glycosylation on ENO1 inhibits glycolysis, and decreases cell proliferation and tumor growth. Blockade of S249 glycosylation on ENO1 reduces PD-L1 expression and enhances T cell-mediated immunity against tumor cells. Notably, elimination of glycosylation at both sites synergizes with PD-L1 monoclonal antibody therapy to promote antitumor immune response. Clinically, ENO1 glycosylation levels are up-regulated and show a positive correlation with PD-L1 levels in human colorectal cancers. Thus, our findings provide a mechanistic understanding of how O-GlcNAcylation bridges aerobic glycolysis and immune evasion to promote tumor growth, suggesting effective therapeutic opportunities.
Collapse
Affiliation(s)
- Qiang Zhu
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
| | - Jingchao Li
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
| | - Haofan Sun
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing100026, China
| | - Zhiya Fan
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing100026, China
| | - Jiating Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Siyuan Chai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Bingyi Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Liming Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Weijie Qin
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing100026, China
| | - Yong Wang
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
| | - Linda C. Hsieh-Wilson
- Division of Chemistry & Chemical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Wen Yi
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| |
Collapse
|
10
|
Yao Q, Hu X, Bian T, Zhang Q, Xue Z, Lv Y, Ren S, Chen Y, Zhang D, Chen L. Role of KLF4 and SIAT7A interaction accelerates myocardial hypertrophy induced by Ang II. J Cell Mol Med 2024; 28:e70144. [PMID: 39431583 PMCID: PMC11492152 DOI: 10.1111/jcmm.70144] [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: 01/02/2024] [Revised: 09/05/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
Sialylation catalysed by sialyltransferase 7A (SIAT7A) plays a role in the development of cardiac hypertrophy. However, the regulatory mechanisms upstream of SIAT7A in this context remain poorly elucidated. Previous study demonstrated that KLF4 activates the SIAT7A gene in ischemic myocardium by binding to its promoter region. Nevertheless, the potential involvement of KLF4 in regulating SIAT7A expression in Ang II-induced hypertrophic cardiomyocytes remains uncertain. This study seeks to deepen the underlying mechanisms of the KLF4 and SIAT7A interaction in the progression of Ang II-induced cardiac hypertrophy. The results showed a concurrent increase in SIAT7A and KLF4 levels in hypertrophic myocardium of essential hypertension patients and in hypertrophic cardiomyocytes stimulated by Ang II. In vitro experiments revealed that reducing KLF4 levels led to a decrease in both SIAT7A synthesis and Sialyl-Tn antigen expression, consequently inhibiting Ang II-induced cardiomyocyte hypertrophy. Intriguingly, reducing SIAT7A levels also resulted in decreased KLF4 expression and suppression cardiomyocyte hypertrophy. Consistent with this, elevating SIAT7A levels increased KLF4 expression and exacerbated cardiomyocyte hypertrophy in both in vivo and in vitro experiments. Additionally, a time-course analysis indicated that KLF4 expression preceded that of SIAT7A. Luciferase reporter assays further confirmed that modulating SIAT7A levels directly influenced the transcriptional activity of KLF4 in cardiomyocytes. In summary, KLF4 expression is upregulated in cardiomyocytes treated with Ang II, which subsequently induces the expression of SIAT7A. The elevated levels of SIAT7A, in turn, enhance the transcription of KLF4. These findings suggest a positive feedback loop between KLF4 and SIAT7A-Sialyl-Tn, ultimately promoting Ang II-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Qiying Yao
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Xinrui Hu
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Tiantian Bian
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Qing Zhang
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Zhao Xue
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yuesheng Lv
- Institute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Shupeng Ren
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yue Chen
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Dongmei Zhang
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Liang Chen
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
11
|
Grilo LF, Zimmerman KD, Puppala S, Chan J, Huber HF, Li G, Jadhav AYL, Wang B, Li C, Clarke GD, Register TC, Oliveira PJ, Nathanielsz PW, Olivier M, Pereira SP, Cox LA. Cardiac Molecular Analysis Reveals Aging-Associated Metabolic Alterations Promoting Glycosaminoglycans Accumulation via Hexosamine Biosynthetic Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309211. [PMID: 39119859 PMCID: PMC11481188 DOI: 10.1002/advs.202309211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Age is a prominent risk factor for cardiometabolic disease, often leading to heart structural and functional changes. However, precise molecular mechanisms underlying cardiac remodeling and dysfunction exclusively resulting from physiological aging remain elusive. Previous research demonstrated age-related functional alterations in baboons, analogous to humans. The goal of this study is to identify early cardiac molecular alterations preceding functional adaptations, shedding light on the regulation of age-associated changes. Unbiased transcriptomics of left ventricle samples are performed from female baboons aged 7.5-22.1 years (human equivalent ≈30-88 years). Weighted-gene correlation network and pathway enrichment analyses are performed, with histological validation. Modules of transcripts negatively correlated with age implicated declined metabolism-oxidative phosphorylation, tricarboxylic acid cycle, glycolysis, and fatty-acid β-oxidation. Transcripts positively correlated with age suggested a metabolic shift toward glucose-dependent anabolic pathways, including hexosamine biosynthetic pathway (HBP). This shift is associated with increased glycosaminoglycan synthesis, modification, precursor synthesis via HBP, and extracellular matrix accumulation, verified histologically. Upregulated extracellular matrix-induced signaling coincided with glycosaminoglycan accumulation, followed by cardiac hypertrophy-related pathways. Overall, these findings revealed a transcriptional shift in metabolism favoring glycosaminoglycan accumulation through HBP before cardiac hypertrophy. Unveiling this metabolic shift provides potential targets for age-related cardiac diseases, offering novel insights into early age-related mechanisms.
Collapse
Affiliation(s)
- Luís F. Grilo
- CNC‐UCCenter for Neuroscience and Cell BiologyUniversity of CoimbraCoimbra3060Portugal
- CIBBCenter for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbra3060Portugal
- Institute for Interdisciplinary ResearchPDBEB – Doctoral Programme in Experimental Biology and BiomedicineUniversity of CoimbraCoimbra3060Portugal
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Kip D. Zimmerman
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Sobha Puppala
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Jeannie Chan
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Hillary F. Huber
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTX78245USA
| | - Ge Li
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Avinash Y. L. Jadhav
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Benlian Wang
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Cun Li
- Texas Pregnancy & Life‐Course Health Research CenterDepartment of Animal ScienceUniversity of WyomingLaramieWY82071USA
| | - Geoffrey D. Clarke
- Department of RadiologyUniversity of Texas Health Science CenterSan AntonioTX78229USA
| | - Thomas C. Register
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Paulo J. Oliveira
- CNC‐UCCenter for Neuroscience and Cell BiologyUniversity of CoimbraCoimbra3060Portugal
- CIBBCenter for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbra3060Portugal
| | - Peter W. Nathanielsz
- Texas Pregnancy & Life‐Course Health Research CenterDepartment of Animal ScienceUniversity of WyomingLaramieWY82071USA
| | - Michael Olivier
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Susana P. Pereira
- CNC‐UCCenter for Neuroscience and Cell BiologyUniversity of CoimbraCoimbra3060Portugal
- CIBBCenter for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbra3060Portugal
- Laboratory of Metabolism and Exercise (LaMetEx)Research Centre in Physical ActivityHealth and Leisure (CIAFEL)Laboratory for Integrative and Translational Research in Population Health (ITR)Faculty of SportsUniversity of PortoPorto4050Portugal
| | - Laura A. Cox
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTX78245USA
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC27157USA
| |
Collapse
|
12
|
Guo B, Zhang F, Yin Y, Ning X, Zhang Z, Meng Q, Yang Z, Jiang W, Liu M, Wang Y, Sun L, Yu L, Mu N. Post-translational modifications of pyruvate dehydrogenase complex in cardiovascular disease. iScience 2024; 27:110633. [PMID: 39224515 PMCID: PMC11367490 DOI: 10.1016/j.isci.2024.110633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Pyruvate dehydrogenase complex (PDC) is a crucial enzyme that connects glycolysis and the tricarboxylic acid (TCA) cycle pathway. It plays an essential role in regulating glucose metabolism for energy production by catalyzing the oxidative decarboxylation of pyruvate to acetyl coenzyme A. Importantly, the activity of PDC is regulated through post-translational modifications (PTMs), phosphorylation, acetylation, and O-GlcNAcylation. These PTMs have significant effects on PDC activity under both physiological and pathophysiological conditions, making them potential targets for metabolism-related diseases. This review specifically focuses on the PTMs of PDC in cardiovascular diseases (CVDs) such as myocardial ischemia/reperfusion injury, diabetic cardiomyopathy, obesity-related cardiomyopathy, heart failure (HF), and vascular diseases. The findings from this review offer theoretical references for the diagnosis, treatment, and prognosis of CVD.
Collapse
Affiliation(s)
- Bo Guo
- Department of Pharmacy, Northwest Woman’s and Children’s Hospital, Xi’an, China
| | - Fujiao Zhang
- College of Life Sciences, Northwest University, Xi’an, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Xingmin Ning
- College of Life Sciences, Northwest University, Xi’an, China
| | - Zihui Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Qinglei Meng
- College of Life Sciences, Yan’an University, Yan’an, China
| | - Ziqi Yang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Lijuan Sun
- Eye Institute of Chinese PLA and Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
13
|
Chiu KY, Ai Y, Tanim-Ai Hassan M, Li X, Gunawardena HP, Chen H. Standards-Free Absolute Quantitation of Oxidizable Glycopeptides by Coulometric Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1441-1450. [PMID: 38815255 DOI: 10.1021/jasms.4c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Currently, glycopeptide quantitation is mainly based on relative quantitation due to absolute quantitation requiring isotope-labeled or standard glycopeptides which may not be commercially available or are very costly and time consuming to synthesize. To address this grand challenge, coulometric mass spectrometry (CMS), based on the combination of electrochemistry (EC) and mass spectrometry (MS), was utilized to quantify electrochemically active glycopeptides without the need of using standard materials. In this study, we studied tyrosine-containing glycopeptides, NYIVGQPSS(β-GlcNAc)TGNL-OH and NYSVPSS(β-GlcNAc)TGNL-OH, and successfully quantified them directly with CMS with a discrepancy of less than 5% between the CMS measured amount and the theoretical amount. Taking one step further, we applied this approach to quantify glycopeptides generated from the digestion of NIST mAb, a monoclonal antibody reference material. Through HILIC column separation, five N297 glycopeptides resulting from NIST mAb tryptic digestion were successfully separated and quantified by CMS for an absolute amount without the use of any standard materials. This study indicates the potential utility of CMS for quantitative proteomics research.
Collapse
Affiliation(s)
- Kai-Yuan Chiu
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Yongling Ai
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Md Tanim-Ai Hassan
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Xuanwen Li
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Harsha P Gunawardena
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Springhouse, Pennsylvania 19002, United States
| | - Hao Chen
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| |
Collapse
|
14
|
Lee S. Cardiovascular Disease and miRNAs: Possible Oxidative Stress-Regulating Roles of miRNAs. Antioxidants (Basel) 2024; 13:656. [PMID: 38929095 PMCID: PMC11200533 DOI: 10.3390/antiox13060656] [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: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) have been highlighted as key players in numerous diseases, and accumulating evidence indicates that pathological expressions of miRNAs contribute to both the development and progression of cardiovascular diseases (CVD), as well. Another important factor affecting the development and progression of CVD is reactive oxygen species (ROS), as well as the oxidative stress they may impose on the cells. Considering miRNAs are involved in virtually every biological process, it is not unreasonable to assume that miRNAs also play critical roles in the regulation of oxidative stress. This narrative review aims to provide mechanistic insights on possible oxidative stress-regulating roles of miRNAs in cardiovascular diseases based on differentially expressed miRNAs reported in various cardiovascular diseases and their empirically validated targets that have been implicated in the regulation of oxidative stress.
Collapse
Affiliation(s)
- Seahyoung Lee
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| |
Collapse
|
15
|
Shi RR, He TQ, Lin MS, Xu J, Gu JH, Xu H. O-GlcNAcylation in ischemic diseases. Front Pharmacol 2024; 15:1377235. [PMID: 38783961 PMCID: PMC11113977 DOI: 10.3389/fphar.2024.1377235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Protein glycosylation is an extensively studied field, with the most studied forms being oxygen or nitrogen-linked N-acetylglucosamine (O-GlcNAc or N-GlcNAc) glycosylation. Particular residues on proteins are targeted by O-GlcNAcylation, which is among the most intricate post-translational modifications. Significantly contributing to an organism's proteome, it influences numerous factors affecting protein stability, function, and subcellular localization. It also modifies the cellular function of target proteins that have crucial responsibilities in controlling pathways related to the central nervous system, cardiovascular homeostasis, and other organ functions. Under conditions of acute stress, changes in the levels of O-GlcNAcylation of these proteins may have a defensive function. Nevertheless, deviant O-GlcNAcylation nullifies this safeguard and stimulates the advancement of several ailments, the prognosis of which relies on the cellular milieu. Hence, this review provides a concise overview of the function and comprehension of O-GlcNAcylation in ischemia diseases, aiming to facilitate the discovery of new therapeutic targets for efficient treatment, particularly in patients with diabetes.
Collapse
Affiliation(s)
- Rui-Rui Shi
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Tian-Qi He
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- Department of Pharmacy, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Meng-Si Lin
- Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Jian Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- Department of Pharmacy, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Jin-Hua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- Department of Pharmacy, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Hui Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| |
Collapse
|
16
|
Vang S, Helton ES, Guo Y, Burpee B, Rose E, Easter M, Bollenbecker S, Hirsch MJ, Matthews EL, Jones LI, Howze PH, Rajasekaran V, Denson R, Cochran P, Attah IK, Olson H, Clair G, Melkani G, Krick S, Barnes JW. O-GlcNAc transferase regulates collagen deposition and fibrosis resolution in idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1387197. [PMID: 38665916 PMCID: PMC11043510 DOI: 10.3389/fimmu.2024.1387197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic pulmonary disease that is characterized by an excessive accumulation of extracellular matrix (ECM) proteins (e.g. collagens) in the parenchyma, which ultimately leads to respiratory failure and death. While current therapies exist to slow the progression, no therapies are available to resolve fibrosis. Methods We characterized the O-linked N-Acetylglucosamine (O-GlcNAc) transferase (OGT)/O-GlcNAc axis in IPF using single-cell RNA-sequencing (scRNA-seq) data and human lung sections and isolated fibroblasts from IPF and non-IPF donors. The underlying mechanism(s) of IPF were further investigated using multiple experimental models to modulate collagen expression and accumulation by genetically and pharmacologically targeting OGT. Furthermore, we hone in on the transforming growth factor-beta (TGF-β) effector molecule, Smad3, by co-expressing it with OGT to determine if it is modified and its subsequent effect on Smad3 activation. Results We found that OGT and O-GlcNAc levels are upregulated in patients with IPF compared to non-IPF. We report that the OGT regulates collagen deposition and fibrosis resolution, which is an evolutionarily conserved process demonstrated across multiple species. Co-expression of OGT and Smad3 showed that Smad3 is O-GlcNAc modified. Blocking OGT activity resulted in decreased phosphorylation at Ser-423/425 of Smad3 attenuating the effects of TGF-β1 induced collagen expression/deposition. Conclusion OGT inhibition or knockdown successfully blocked and reversed collagen expression and accumulation, respectively. Smad3 is discovered to be a substrate of OGT and its O-GlcNAc modification(s) directly affects its phosphorylation state. These data identify OGT as a potential target in pulmonary fibrosis resolution, as well as other diseases that might have aberrant ECM/collagen accumulation.
Collapse
Affiliation(s)
- Shia Vang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eric Scott Helton
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yiming Guo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bailey Burpee
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Elex Rose
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Molly Easter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Seth Bollenbecker
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meghan June Hirsch
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Emma Lea Matthews
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Luke Isaac Jones
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Patrick Henry Howze
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vasanthi Rajasekaran
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rebecca Denson
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Phillip Cochran
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Isaac Kwame Attah
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Heather Olson
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Girish Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stefanie Krick
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jarrod Wesley Barnes
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
17
|
Jagannath S, Mallanna SH, Nandini CD. Diet-inducing hypercholesterolemia show decreased O-GlcNAcylation of liver proteins through modulation of AMPK. J Physiol Biochem 2024; 80:205-218. [PMID: 37996652 DOI: 10.1007/s13105-023-00997-7] [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: 04/19/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
O-GlcNAcylation, a nutritionally driven, post-translational modification of proteins, is gaining importance because of its health implications. Changes in O-GlcNAcylation are observed in various disease conditions. Changes in O-GlcNAcylation by diet that causes hypercholesterolemia are not critically looked into in the liver. To address it, both in vitro and in vivo approaches were employed. Hypercholesterolemia was induced individually by feeding cholesterol (H)/high-fat (HF) diet. Global O-GlcNAcylation levels and modulation of AMPK activation in both preventive and curative approaches were looked into. Diet-induced hypercholesterolemia resulted in decreased O-GlcNAcylation of liver proteins which was associated with decreased O-linked N-acetylglucosaminyltransferase (OGT) and Glutamine fructose-6-phosphate amidotransferase-1 (GFAT1). Activation of AMPK by metformin in preventive mode restored the O-GlcNAcylation levels; however, metformin treatment of HepG2 cells in curative mode restored O-GlcNAcylation levels in HF but failed to in H condition (at 24 h). Further, maternal faulty diet resulted in decreased O-GlcNAcylation in pup liver despite feeding normal diet till adulthood. A faulty diet modulates global O-GlcNAcylation of liver proteins which is accompanied by decreased AMPK activation which could exacerbate metabolic syndromes through fat accumulation in the liver.
Collapse
Affiliation(s)
- Sanjana Jagannath
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Smitha Honnalagere Mallanna
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - C D Nandini
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, 570020, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
18
|
Costa TJ, Wilson EW, Fontes MT, Pernomian L, Tostes RC, Wenceslau CF, McCarthy CG. The O-GlcNAc dichotomy: when does adaptation become pathological? Clin Sci (Lond) 2023; 137:1683-1697. [PMID: 37986614 DOI: 10.1042/cs20220309] [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: 07/13/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023]
Abstract
O-Linked attachment of β-N-acetylglucosamine (O-GlcNAc) on serine and threonine residues of nuclear, cytoplasmic, and mitochondrial proteins is a highly dynamic and ubiquitous post-translational modification that impacts the function, activity, subcellular localization, and stability of target proteins. Physiologically, acute O-GlcNAcylation serves primarily to modulate cellular signaling and transcription regulatory pathways in response to nutrients and stress. To date, thousands of proteins have been revealed to be O-GlcNAcylated and this number continues to grow as the technology for the detection of O-GlcNAc improves. The attachment of a single O-GlcNAc is catalyzed by the enzyme O-GlcNAc transferase (OGT), and their removal is catalyzed by O-GlcNAcase (OGA). O-GlcNAcylation is regulated by the metabolism of glucose via the hexosamine biosynthesis pathway, and the metabolic abnormalities associated with pathophysiological conditions are all associated with increased flux through this pathway and elevate O-GlcNAc levels. While chronic O-GlcNAcylation is well associated with cardiovascular dysfunction, only until recently, and with genetically modified animals, has O-GlcNAcylation as a contributing mechanism of cardiovascular disease emerged. This review will address and critically evaluate the current literature on the role of O-GlcNAcylation in vascular physiology, with a view that this pathway can offer novel targets for the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Tiago J Costa
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC, U.S.A
| | - Emily W Wilson
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, SC, U.S.A
| | - Milene T Fontes
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC, U.S.A
| | - Laena Pernomian
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC, U.S.A
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Camilla F Wenceslau
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC, U.S.A
| | - Cameron G McCarthy
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, SC, U.S.A
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC, U.S.A
| |
Collapse
|
19
|
Grilo LF, Zimmerman KD, Puppala S, Chan J, Huber HF, Li G, Jadhav AYL, Wang B, Li C, Clarke GD, Register TC, Oliveira PJ, Nathanielsz PW, Olivier M, Pereira SP, Cox LA. Cardiac Molecular Analysis Reveals Aging-Associated Metabolic Alterations Promoting Glycosaminoglycans Accumulation Via Hexosamine Biosynthetic Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567640. [PMID: 38014295 PMCID: PMC10680868 DOI: 10.1101/2023.11.17.567640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Age is a prominent risk factor for cardiometabolic disease, and often leads to heart structural and functional changes. However, precise molecular mechanisms underlying cardiac remodeling and dysfunction resulting from physiological aging per se remain elusive. Understanding these mechanisms requires biological models with optimal translation to humans. Previous research demonstrated that baboons undergo age-related reduction in ejection fraction and increased heart sphericity, mirroring changes observed in humans. The goal of this study was to identify early cardiac molecular alterations that precede functional adaptations, shedding light on the regulation of age-associated changes. We performed unbiased transcriptomics of left ventricle (LV) samples from female baboons aged 7.5-22.1 years (human equivalent ~30-88 years). Weighted-gene correlation network and pathway enrichment analyses were performed to identify potential age-associated mechanisms in LV, with histological validation. Myocardial modules of transcripts negatively associated with age were primarily enriched for cardiac metabolism, including oxidative phosphorylation, tricarboxylic acid cycle, glycolysis, and fatty-acid β-oxidation. Transcripts positively correlated with age suggest upregulation of glucose uptake, pentose phosphate pathway, and hexosamine biosynthetic pathway (HBP), indicating a metabolic shift towards glucose-dependent anabolic pathways. Upregulation of HBP commonly results in increased glycosaminoglycan precursor synthesis. Transcripts involved in glycosaminoglycan synthesis, modification, and intermediate metabolism were also upregulated in older animals, while glycosaminoglycan degradation transcripts were downregulated with age. These alterations would promote glycosaminoglycan accumulation, which was verified histologically. Upregulation of extracellular matrix (ECM)-induced signaling pathways temporally coincided with glycosaminoglycan accumulation. We found a subsequent upregulation of cardiac hypertrophy-related pathways and an increase in cardiomyocyte width. Overall, our findings revealed a transcriptional shift in metabolism from catabolic to anabolic pathways that leads to ECM glycosaminoglycan accumulation through HBP prior to upregulation of transcripts of cardiac hypertrophy-related pathways. This study illuminates cellular mechanisms that precede development of cardiac hypertrophy, providing novel potential targets to remediate age-related cardiac diseases.
Collapse
Affiliation(s)
- Luís F. Grilo
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
- University of Coimbra, Institute for Interdisciplinary Research, PDBEB - Doctoral Programme in Experimental Biology and Biomedicine
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Kip D. Zimmerman
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sobha Puppala
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jeannie Chan
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Hillary F. Huber
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ge Li
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Avinash Y. L. Jadhav
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Benlian Wang
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Cun Li
- Texas Pregnancy & Life-Course Health Research Center, Department of Animal Science, University of Wyoming, Laramie, Wyoming, USA
| | - Geoffrey D. Clarke
- Department of Radiology, University of Texas Health Science Center, San Antonio, Texas
| | - Thomas C. Register
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Paulo J. Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Peter W. Nathanielsz
- Texas Pregnancy & Life-Course Health Research Center, Department of Animal Science, University of Wyoming, Laramie, Wyoming, USA
| | - Michael Olivier
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Susana P. Pereira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Laura A. Cox
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
20
|
Ednie AR, Paul-Onyia CD, Bennett ES. Reduced O-GlcNAcylation diminishes cardiomyocyte Ca 2+ dependent facilitation and frequency dependent acceleration of relaxation. J Mol Cell Cardiol 2023; 180:10-21. [PMID: 37120927 DOI: 10.1016/j.yjmcc.2023.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Ca2+ dependent facilitation (CDF) and frequency dependent acceleration of relaxation (FDAR) are regulatory mechanisms that potentiate cardiomyocyte Ca2+ channel function and increase the rate of Ca2+ sequestration following a Ca2+-release event, respectively, when depolarization frequency increases. CDF and FDAR likely evolved to maintain EC coupling at increased heart rates. Ca2+/calmodulin-dependent kinase II (CaMKII) was shown to be indispensable to both; however, the mechanisms remain to be completely elucidated. CaMKII activity can be modulated by post-translational modifications but if and how these modifications impact CDF and FDAR is unknown. Intracellular O-linked glycosylation (O-GlcNAcylation) is a post-translational modification that acts as a signaling molecule and metabolic sensor. In hyperglycemic conditions, CaMKII was shown to be O-GlcNAcylated resulting in pathologic activity. Here we sought to investigate whether O-GlcNAcylation impacts CDF and FDAR through modulation of CaMKII activity in a pseudo-physiologic setting. Using voltage-clamp and Ca2+ photometry we show that cardiomyocyte CDF and FDAR are significantly diminished in conditions of reduced O-GlcNAcylation. Immunoblot showed that CaMKIIδ and calmodulin expression are increased but the autophosphorylation of CaMKIIδ and the muscle cell-specific CaMKIIβ isoform are reduced by 75% or more when O-GlcNAcylation is inhibited. We also show that the enzyme responsible for O-GlcNAcylation (OGT) can likely be localized in the dyad space and/or at the cardiac sarcoplasmic reticulum and is precipitated by calmodulin in a Ca2+ dependent manner. These findings will have important implications for our understanding of how CaMKII and OGT interact to impact cardiomyocyte EC coupling in normal physiologic settings as well as in disease states where CaMKII and OGT may be aberrantly regulated.
Collapse
Affiliation(s)
- Andrew R Ednie
- Department of Neuroscience, Cell Biology & Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, USA.
| | - Chiagozie D Paul-Onyia
- Department of Neuroscience, Cell Biology & Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, USA
| | - Eric S Bennett
- Department of Neuroscience, Cell Biology & Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, USA
| |
Collapse
|
21
|
Zhang L, Li J, Chen J, Lei J, Yuan Z, Zhang J, Liu Z, Yu C, Ma L. Oscillatory shear stress-mediated aberrant O-GlcNAc SIRT3 accelerates glycocalyx inflammatory injury via LKB1/p47 phox/Hyal2 signaling. Cell Signal 2023:110790. [PMID: 37392860 DOI: 10.1016/j.cellsig.2023.110790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/07/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Glycocalyx coating on endothelial surface layer helps to sense shear forces and maintain endothelial function. However, the underlying mechanism of endothelial glycocalyx degradation upon disordered shear stress stimulation is not fully understood. SIRT3, a major NAD+-dependent protein deacetylases, is required for protein stability during vascular homeostasis and partly involved in atherosclerotic process. While few studies showed that SIRT3 is responsible for endothelial glycocalyx homeostasis under shear stress, the underlying mechanisms remain largely unknown. Here, we demonstrated that oscillatory shear stress (OSS) induces glycocalyx injury by activating LKB1/p47phox/Hyal2 axis both in vivo and in vitro. And O-GlcNAc modification served to prolong SIRT3 deacetylase activity and stabilized p47/Hyal2 complex. OSS could decrease SIRT3 O-GlcNAcylation to activate LKB1, further accelerated endothelial glycocalyx injury in inflammatory microenvironment. SIRT3Ser329 mutation or inhibition of SIRT3 O-GlcNAcylation strongly promoted glycocalyx degradation. On the contrary, overexpression of SIRT3 reverse glycocalyx damage upon OSS treatment. Together, our findings indicated that targeting O-GlcNAcylation of SIRT3 could prevent and/or treat diseases whereby glycocalyx injured.
Collapse
Affiliation(s)
- Lei Zhang
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jiajia Li
- Hechuan District People's Hospital, Chongqing, China
| | - Jun Chen
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jin Lei
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zhiyi Yuan
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Zhaohong Liu
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chao Yu
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China.
| | - Limei Ma
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
22
|
Khanal S, Bhavnani N, Mathias A, Lallo J, Gupta S, Ohanyan V, Ferrell JM, Raman P. Deletion of Smooth Muscle O-GlcNAc Transferase Prevents Development of Atherosclerosis in Western Diet-Fed Hyperglycemic ApoE -/- Mice In Vivo. Int J Mol Sci 2023; 24:7899. [PMID: 37175604 PMCID: PMC10178779 DOI: 10.3390/ijms24097899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Accumulating evidence highlights protein O-GlcNAcylation as a putative pathogenic contributor of diabetic vascular complications. We previously reported that elevated protein O-GlcNAcylation correlates with increased atherosclerotic lesion formation and VSMC proliferation in response to hyperglycemia. However, the role of O-GlcNAc transferase (OGT), regulator of O-GlcNAc signaling, in the evolution of diabetic atherosclerosis remains elusive. The goal of this study was to determine whether smooth muscle OGT (smOGT) plays a direct role in hyperglycemia-induced atherosclerotic lesion formation and SMC de-differentiation. Using tamoxifen-inducible Myh11-CreERT2 and Ogtfl/fl mice, we generated smOGTWT and smOGTKO mice, with and without ApoE-null backgrounds. Following STZ-induced hyperglycemia, smOGTWT and smOGTKO mice were kept on a standard laboratory diet for the study duration. In a parallel study, smOGTWTApoE-/- and smOGTKOApoE-/- were initiated on Western diet at 8-wks-age. Animals harvested at 14-16-wks-age were used for plasma and tissue collection. Loss of smOGT augmented SM contractile marker expression in aortic vessels of STZ-induced hyperglycemic smOGTKO mice. Consistently, smOGT deletion attenuated atherosclerotic lesion lipid burden (Oil red O), plaque area (H&E), leukocyte (CD45) and smooth muscle cell (ACTA2) abundance in Western diet-fed hyperglycemic smOGTKOApoE-/- mice. This was accompanied by increased SM contractile markers and reduced inflammatory and proliferative marker expression. Further, smOGT deletion attenuated YY1 and SRF expression (transcriptional regulators of SM contractile genes) in hyperglycemic smOGTKOApoE-/- and smOGTKO mice. These data uncover an athero-protective outcome of smOGT loss-of-function and suggest a direct regulatory role of OGT-mediated O-GlcNAcylation in VSMC de-differentiation in hyperglycemia.
Collapse
Affiliation(s)
- Saugat Khanal
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Neha Bhavnani
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Amy Mathias
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
| | - Jason Lallo
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
| | - Shreya Gupta
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Jessica M. Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Priya Raman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (S.K.)
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
23
|
Wu K, Chen L, Qiu Z, Zhao B, Hou J, Lei S, Jiang M, Xia Z. Protective Effect and Mechanism of Xbp1s Regulating HBP/O-GlcNAcylation through GFAT1 on Brain Injury after SAH. Biomedicines 2023; 11:biomedicines11051259. [PMID: 37238930 DOI: 10.3390/biomedicines11051259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
(1) SAH induces cellular stress and endoplasmic reticulum stress, activating the unfolded protein response (UPR) in nerve cells. IRE1 (inositol-requiring enzyme 1) is a protein that plays a critical role in cellular stress response. Its final product, Xbp1s, is essential for adapting to changes in the external environment. This process helps maintain proper cellular function in response to various stressors. O-GlcNAcylation, a means of protein modification, has been found to be involved in SAH pathophysiology. SAH can increase the acute O-GlcNAcylation level of nerve cells, which enhances the stress capacity of nerve cells. The GFAT1 enzyme regulates the level of O-GlcNAc modification in cells, which could be a potential target for neuroprotection in SAH. Investigating the IRE1/XBP1s/GFAT1 axis could offer a promising avenue for future research. (2) Methods: SAH was induced using a suture to perforate an artery in mice. HT22 cells with Xbp1 loss- and gain-of-function in neurons were generated. Thiamet-G was used to increase O-GlcNAcylation; (3) Results: Severe neuroinflammation caused by subarachnoid hemorrhage leads to extensive endoplasmic reticulum stress of nerve cells. Xbp1s, the final product of unfolded proteins induced by endoplasmic reticulum stress, can induce the expression of the hexosamine pathway rate limiting enzyme GFAT1, increase the level of O-GlcNAc modification of cells, and have a protective effect on neural cells; (4) Conclusions: The correlation between Xbp1s displayed by immunohistochemistry and O-GlcNAc modification suggests that the IRE1/XBP1 branch of unfolded protein reaction plays a key role in subarachnoid hemorrhage. IRE1/XBP1 branch is a new idea to regulate protein glycosylation modification, and provides a promising strategy for clinical perioperative prevention and treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Kefan Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Lili Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Shaoqin Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Meng Jiang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| |
Collapse
|
24
|
Papanicolaou KN, Jung J, Ashok D, Zhang W, Modaressanavi A, Avila E, Foster DB, Zachara NE, O'Rourke B. Inhibiting O-GlcNAcylation impacts p38 and Erk1/2 signaling and perturbs cardiomyocyte hypertrophy. J Biol Chem 2023; 299:102907. [PMID: 36642184 PMCID: PMC9988579 DOI: 10.1016/j.jbc.2023.102907] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The dynamic cycling of O-linked GlcNAc (O-GlcNAc) on and off Ser/Thr residues of intracellular proteins, termed O-GlcNAcylation, is mediated by the conserved enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase. O-GlcNAc cycling is important in homeostatic and stress responses, and its perturbation sensitizes the heart to ischemic and other injuries. Despite considerable progress, many molecular pathways impacted by O-GlcNAcylation in the heart remain unclear. The mitogen-activated protein kinase (MAPK) pathway is a central signaling cascade that coordinates developmental, physiological, and pathological responses in the heart. The developmental or adaptive arm of MAPK signaling is primarily mediated by Erk kinases, while the pathophysiologic arm is mediated by p38 and Jnk kinases. Here, we examine whether O-GlcNAcylation affects MAPK signaling in cardiac myocytes, focusing on Erk1/2 and p38 in basal and hypertrophic conditions induced by phenylephrine. Using metabolic labeling of glycans coupled with alkyne-azide "click" chemistry, we found that Erk1/2 and p38 are O-GlcNAcylated. Supporting the regulation of p38 by O-GlcNAcylation, the OGT inhibitor, OSMI-1, triggers the phosphorylation of p38, an event that involves the NOX2-Ask1-MKK3/6 signaling axis and also the noncanonical activator Tab1. Additionally, OGT inhibition blocks the phenylephrine-induced phosphorylation of Erk1/2. Consistent with perturbed MAPK signaling, OSMI-1-treated cardiomyocytes have a blunted hypertrophic response to phenylephrine, decreased expression of cTnT (key component of the contractile apparatus), and increased expression of maladaptive natriuretic factors Anp and Bnp. Collectively, these studies highlight new roles for O-GlcNAcylation in maintaining a balanced activity of Erk1/2 and p38 MAPKs during hypertrophic growth responses in cardiomyocytes.
Collapse
Affiliation(s)
- Kyriakos N Papanicolaou
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Jessica Jung
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Deepthi Ashok
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wenxi Zhang
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Modaressanavi
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eddie Avila
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - D Brian Foster
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natasha E Zachara
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
25
|
Escobar EE, Seeley EH, Serrano-Negrón JE, Vocadlo DJ, Brodbelt JS. In Situ Imaging of O-Linked β-N-Acetylglucosamine Using On-Tissue Hydrolysis and MALDI Mass Spectrometry. Cancers (Basel) 2023; 15:1224. [PMID: 36831567 PMCID: PMC9954453 DOI: 10.3390/cancers15041224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Post-translational O-glycosylation of proteins via the addition of N-acetylglucosamine (O-GlcNAc) is a regulator of many aspects of cellular physiology. Processes driven by perturbed dynamics of O-GlcNAcylation modification have been implicated in cancer development. Variability in O-GlcNAcylation is emerging as a metabolic biomarker of many cancers. Here, we evaluate the use of MALDI-mass spectrometry imaging (MSI) to visualize the location of O-GlcNAcylated proteins in tissue sections by mapping GlcNAc that has been released by the enzymatic hydrolysis of glycoproteins using an O-GlcNAc hydrolase. We use this strategy to monitor O-GlcNAc within hepatic VX2 tumor tissue. We show that increased O-GlcNAc is found within both viable tumor and tumor margin regions, implicating GlcNAc in tumor progression.
Collapse
Affiliation(s)
- Edwin E. Escobar
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | - Erin H. Seeley
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | | | - David J. Vocadlo
- Department of Molecular Biology and Biochemistry, Burnaby, BC V5A 1S6, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Jennifer S. Brodbelt
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
26
|
Wang Y, Fang X, Wang S, Wang B, Chu F, Tian Z, Zhang L, Zhou F. The role of O-GlcNAcylation in innate immunity and inflammation. J Mol Cell Biol 2023; 14:6880149. [PMID: 36473120 PMCID: PMC9951266 DOI: 10.1093/jmcb/mjac065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/30/2022] [Accepted: 09/06/2022] [Indexed: 12/12/2022] Open
Abstract
O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) is a highly dynamic and widespread post-translational modification (PTM) that regulates the activity, subcellular localization, and stability of target proteins. O-GlcNAcylation is a reversible PTM controlled by two cycling enzymes: O-linked N-acetylglucosamine transferase and O-GlcNAcase. Emerging evidence indicates that O-GlcNAcylation plays critical roles in innate immunity, inflammatory signaling, and cancer development. O-GlcNAcylation usually occurs on serine/threonine residues, where it interacts with other PTMs, such as phosphorylation. Thus, it likely has a broad regulatory scope. This review discusses the recent research advances regarding the regulatory roles of O-GlcNAcylation in innate immunity and inflammation. A more comprehensive understanding of O-GlcNAcylation could help to optimize therapeutic strategies regarding inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Yongqiang Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Xiuwu Fang
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Shuai Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Bin Wang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Feng Chu
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Zhixin Tian
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
27
|
Matsuno M, Yokoe S, Nagatsuka T, Morihara H, Moriwaki K, Asahi M. O-GlcNAcylation-induced GSK-3β activation deteriorates pressure overload-induced heart failure via lack of compensatory cardiac hypertrophy in mice. Front Endocrinol (Lausanne) 2023; 14:1122125. [PMID: 37033243 PMCID: PMC10073727 DOI: 10.3389/fendo.2023.1122125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
O-GlcNAc transferase (OGT) modulates many functions of proteins via O-GlcNAcylation that adds O-linked β-N-acetylglucosamine (O-GlcNAc) to the serine/threonine residues of proteins. However, the role of O-GlcNAcylation in cardiac remodeling and function is not fully understood. To examine the effect of O-GlcNAcylation on pressure overload-induced cardiac hypertrophy and subsequent heart failure, transverse aortic constriction (TAC) surgery was performed in wild type (WT) and Ogt transgenic (Ogt-Tg) mice. Four weeks after TAC (TAC4W), the heart function of Ogt-Tg mice was significantly lower than that of WT mice (reduced fractional shortening and increased ANP levels). The myocardium of left ventricle (LV) in Ogt-Tg mice became much thinner than that in WT mice. Moreover, compared to the heart tissues of WT mice, O-GlcNAcylation of GSK-3β at Ser9 was increased and phosphorylation of GSK-3β at Ser9 was reduced in the heart tissues of Ogt-Tg mice, resulting in its activation and subsequent inactivation of nuclear factor of activated T cell (NFAT) activity. Finally, the thinned LV wall and reduced cardiac function induced by TAC4W in Ogt-Tg mice was reversed by the treatment of a GSK-3β inhibitor, TDZD-8. These results imply that augmented O-GlcNAcylation exacerbates pressure overload-induced heart failure due to a lack of compensatory cardiac hypertrophy via O-GlcNAcylation of GSK-3β, which deprives the phosphorylation site of GSK-3β to constantly inactivate NFAT activity to prevent cardiac hypertrophy. Our findings may provide a new therapeutic strategy for cardiac hypertrophy and subsequent heart failure.
Collapse
Affiliation(s)
- Mahito Matsuno
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shunichi Yokoe
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takehiro Nagatsuka
- Center for Medical Research & Development, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Hirofumi Morihara
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kazumasa Moriwaki
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
- *Correspondence: Michio Asahi,
| |
Collapse
|
28
|
Young ME, Latimer MN. Circadian rhythms in cardiac metabolic flexibility. Chronobiol Int 2023; 40:13-26. [PMID: 34162286 PMCID: PMC8695643 DOI: 10.1080/07420528.2021.1939366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022]
Abstract
Numerous aspects of cardiovascular physiology (e.g., heart rate, blood pressure) and pathology (e.g., myocardial infarction and sudden cardiac death) exhibit time-of-day-dependency. In association with day-night differences in energetic demand and substrate availability, the healthy heart displays remarkable metabolic flexibility through temporal partitioning of the metabolic fate of common substrates (glucose, lipid, amino acids). The purpose of this review is to highlight the contribution that circadian clocks provide toward 24-hr fluctuations in cardiac metabolism and to discuss whether attenuation and/or augmentation of these metabolic rhythms through adjustment of nutrient intake timing impacts cardiovascular disease development.
Collapse
Affiliation(s)
- Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - Mary N Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| |
Collapse
|
29
|
Zhou Y, Li Z, Xu M, Zhang D, Ling J, Yu P, Shen Y. O-GlycNacylation Remission Retards the Progression of Non-Alcoholic Fatty Liver Disease. Cells 2022; 11:cells11223637. [PMID: 36429065 PMCID: PMC9688300 DOI: 10.3390/cells11223637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic disease spectrum associated with insulin resistance (IR), from non-alcoholic fatty liver (NAFL) to non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). O-GlcNAcylation is a posttranslational modification, regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Abnormal O-GlcNAcylation plays a key role in IR, fat deposition, inflammatory injury, fibrosis, and tumorigenesis. However, the specific mechanisms and clinical treatments of O-GlcNAcylation and NAFLD are yet to be elucidated. The modification contributes to understanding the pathogenesis and development of NAFLD, thus clarifying the protective effect of O-GlcNAcylation inhibition on liver injury. In this review, the crucial role of O-GlcNAcylation in NAFLD (from NAFL to HCC) is discussed, and the effect of therapeutics on O-GlcNAcylation and its potential mechanisms on NAFLD have been highlighted. These inferences present novel insights into the pathogenesis and treatments of NAFLD.
Collapse
Affiliation(s)
- Yicheng Zhou
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Branch of Nationlal Clinical Research Center for Metabolic Diseases, Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, Nanchang 330031, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Branch of Nationlal Clinical Research Center for Metabolic Diseases, Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong
| | - Jitao Ling
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Branch of Nationlal Clinical Research Center for Metabolic Diseases, Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Branch of Nationlal Clinical Research Center for Metabolic Diseases, Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
- Correspondence: (P.Y.); (Y.S.)
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Branch of Nationlal Clinical Research Center for Metabolic Diseases, Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
- Correspondence: (P.Y.); (Y.S.)
| |
Collapse
|
30
|
Zhu WZ, Palazzo T, Zhou M, Ledee D, Olson HM, Paša-Tolić L, Olson AK. First comprehensive identification of cardiac proteins with putative increased O-GlcNAc levels during pressure overload hypertrophy. PLoS One 2022; 17:e0276285. [PMID: 36288343 PMCID: PMC9605332 DOI: 10.1371/journal.pone.0276285] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022] Open
Abstract
Protein posttranslational modifications (PTMs) by O-GlcNAc globally rise during pressure-overload hypertrophy (POH). However, a major knowledge gap exists on the specific proteins undergoing changes in O-GlcNAc levels during POH primarily because this PTM is low abundance and easily lost during standard mass spectrometry (MS) conditions used for protein identification. Methodologies have emerged to enrich samples for O-GlcNAcylated proteins prior to MS analysis. Accordingly, our goal was to identify the specific proteins undergoing changes in O-GlcNAc levels during POH. We used C57/Bl6 mice subjected to Sham or transverse aortic constriction (TAC) to create POH. From the hearts, we labelled the O-GlcNAc moiety with tetramethylrhodamine azide (TAMRA) before sample enrichment by TAMRA immunoprecipitation (IP). We used LC-MS/MS to identify and quantify the captured putative O-GlcNAcylated proteins. We identified a total of 700 putative O-GlcNAcylated proteins in Sham and POH. Two hundred thirty-three of these proteins had significantly increased enrichment in POH over Sham suggesting higher O-GlcNAc levels whereas no proteins were significantly decreased by POH. We examined two MS identified metabolic enzymes, CPT1B and the PDH complex, to validate by immunoprecipitation. We corroborated increased O-GlcNAc levels during POH for CPT1B and the PDH complex. Enzyme activity assays suggests higher O-GlcNAcylation increases CPT1 activity and decreases PDH activity during POH. In summary, we generated the first comprehensive list of proteins with putative changes in O-GlcNAc levels during POH. Our results demonstrate the large number of potential proteins and cellular processes affected by O-GlcNAc and serve as a guide for testing specific O-GlcNAc-regulated mechanisms during POH.
Collapse
Affiliation(s)
- Wei Zhong Zhu
- Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Teresa Palazzo
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Mowei Zhou
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Dolena Ledee
- Seattle Children’s Research Institute, Seattle, Washington, United States of America,Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Heather M. Olson
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Ljiljana Paša-Tolić
- Environmental Molecular Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Aaron K. Olson
- Seattle Children’s Research Institute, Seattle, Washington, United States of America,Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America,* E-mail:
| |
Collapse
|
31
|
Jo S, Pritchard S, Wong A, Avula N, Essawy A, Hanover J, Alejandro EU. Pancreatic β-cell hyper-O-GlcNAcylation leads to impaired glucose homeostasis in vivo. Front Endocrinol (Lausanne) 2022; 13:1040014. [PMID: 36387851 PMCID: PMC9644030 DOI: 10.3389/fendo.2022.1040014] [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: 09/08/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Protein O-GlcNAcylation is a nutrient and stress-sensitive protein post-translational modification (PTM). The addition of an O-GlcNAc molecule to proteins is catalyzed by O-GlcNAc transferase (OGT), whereas O-GlcNAcase (OGA) enzyme is responsible for removal of this PTM. Previous work showed that OGT is highly expressed in the pancreas, and we demonstrated that hypo-O-GlcNAcylation in β-cells cause severe diabetes in mice. These studies show a direct link between nutrient-sensitive OGT and β-cell health and function. In the current study, we hypothesized that hyper-O-GlcNAcylation may confer protection from β-cell failure in high-fat diet (HFD)-induced obesity. To test this hypothesis, we generated a mouse model with constitutive β-cell OGA ablation (βOGAKO) to specifically increase O-GlcNAcylation in β-cells. Under normal chow diet, young male and female βOGAKO mice exhibited normal glucose tolerance but developed glucose intolerance with aging, relative to littermate controls. No alteration in β-cell mass was observed between βOGAKO and littermate controls. Total insulin content was reduced despite an increase in pro-insulin to insulin ratio in βOGAKO islets. βOGAKO mice showed deficit in insulin secretion in vivo and in vitro. When young animals were subjected to HFD, both male and female βOGAKO mice displayed normal body weight gain and insulin tolerance but developed glucose intolerance that worsened with longer exposure to HFD. Comparable β-cell mass was found between βOGAKO and littermate controls. Taken together, these data demonstrate that the loss of OGA in β-cells reduces β-cell function, thereby perturbing glucose homeostasis. The findings reinforce the rheostat model of intracellular O-GlcNAcylation where too much (OGA loss) or too little (OGT loss) O-GlcNAcylation are both detrimental to the β-cell.
Collapse
Affiliation(s)
- Seokwon Jo
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Samantha Pritchard
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Alicia Wong
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, United States
| | - Nandini Avula
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Ahmad Essawy
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - John Hanover
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Emilyn U. Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
32
|
Zhou F, Ma J, Zhu Y, Wang T, Yang Y, Sun Y, Chen Y, Song H, Huo X, Zhang J. The role and potential mechanism of O-Glycosylation in gastrointestinal tumors. Pharmacol Res 2022; 184:106420. [PMID: 36049664 DOI: 10.1016/j.phrs.2022.106420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/15/2022] [Accepted: 08/26/2022] [Indexed: 10/15/2022]
Abstract
Glycosylation is a critical post-translational modification (PTM) that affects the function of proteins and regulates cell signaling, thereby regulating various biological processes. Protein oxygen-N-acetylglucosamine (O-GlcNAc) glycosylation modifications are glycochemical modifications that occur within cells in the signal transduction and are frequently found in the cytoplasm and nucleus. Due to the rapid and reversible addition and removal, O-GlcNAc modifications are able to reversibly compete with certain phosphorylation modifications, immediately regulate the activity of proteins, and participate in kinds of cellular metabolic and signal transduction pathways, playing a pivotal role in the regulation of tumors, diabetes, and other diseases. This article provided a brief overview of O-GlcNAc glycosylation modification, introduced its role in altering the progression and immune response regulation of gastrointestinal tumors, and discussed its potential use as a marker of tumor neogenesis.
Collapse
Affiliation(s)
- Feinan Zhou
- The department of Spleen and Stomach Diseases of Cadres Healthcare Centre, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Jia Ma
- The First Department of Oncology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Yongfu Zhu
- The First Department of Oncology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Tianming Wang
- Laboratory of Infection and Immunity, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Yue Yang
- Laboratory of Infection and Immunity, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Yehan Sun
- The First Department of Oncology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Youmou Chen
- The First Department of Oncology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Hang Song
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Xingxing Huo
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui Province 230000, China.
| | - Jianye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong Province 510799, China.
| |
Collapse
|
33
|
Silva-Aguiar RP, Peruchetti DB, Pinheiro AAS, Caruso-Neves C, Dias WB. O-GlcNAcylation in Renal (Patho)Physiology. Int J Mol Sci 2022; 23:ijms231911260. [PMID: 36232558 PMCID: PMC9569498 DOI: 10.3390/ijms231911260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/29/2022] Open
Abstract
Kidneys maintain internal milieu homeostasis through a well-regulated manipulation of body fluid composition. This task is performed by the correlation between structure and function in the nephron. Kidney diseases are chronic conditions impacting healthcare programs globally, and despite efforts, therapeutic options for its treatment are limited. The development of chronic degenerative diseases is associated with changes in protein O-GlcNAcylation, a post-translation modification involved in the regulation of diverse cell function. O-GlcNAcylation is regulated by the enzymatic balance between O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) which add and remove GlcNAc residues on target proteins, respectively. Furthermore, the hexosamine biosynthetic pathway provides the substrate for protein O-GlcNAcylation. Beyond its physiological role, several reports indicate the participation of protein O-GlcNAcylation in cardiovascular, neurodegenerative, and metabolic diseases. In this review, we discuss the impact of protein O-GlcNAcylation on physiological renal function, disease conditions, and possible future directions in the field.
Collapse
Affiliation(s)
- Rodrigo P. Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Diogo B. Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Ana Acacia S. Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro 21045-900, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro 21045-900, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Wagner B. Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Correspondence:
| |
Collapse
|
34
|
Azam T, Zhang H, Zhou F, Wang X. Recent Advances on Drug Development and Emerging Therapeutic Agents Through Targeting Cellular Homeostasis for Ageing and Cardiovascular Disease. FRONTIERS IN AGING 2022; 3:888190. [PMID: 35821839 PMCID: PMC9261412 DOI: 10.3389/fragi.2022.888190] [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] [Received: 03/02/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Ageing is a progressive physiological process mediated by changes in biological pathways, resulting in a decline in tissue and cellular function. It is a driving factor in numerous age-related diseases including cardiovascular diseases (CVDs). Cardiomyopathies, hypertension, ischaemic heart disease, and heart failure are some of the age-related CVDs that are the leading causes of death worldwide. Although individual CVDs have distinct clinical and pathophysiological manifestations, a disturbance in cellular homeostasis underlies the majority of diseases which is further compounded with aging. Three key evolutionary conserved signalling pathways, namely, autophagy, mitophagy and the unfolded protein response (UPR) are involved in eliminating damaged and dysfunctional organelle, misfolded proteins, lipids and nucleic acids, together these molecular processes protect and preserve cellular homeostasis. However, amongst the numerous molecular changes during ageing, a decline in the signalling of these key molecular processes occurs. This decline also increases the susceptibility of damage following a stressful insult, promoting the development and pathogenesis of CVDs. In this review, we discuss the role of autophagy, mitophagy and UPR signalling with respect to ageing and cardiac disease. We also highlight potential therapeutic strategies aimed at restoring/rebalancing autophagy and UPR signalling to maintain cellular homeostasis, thus mitigating the pathological effects of ageing and CVDs. Finally, we highlight some limitations that are likely hindering scientific drug research in this field.
Collapse
Affiliation(s)
- Tayyiba Azam
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Hongyuan Zhang
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Fangchao Zhou
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Xin Wang
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
35
|
Fan X, Song Q, Sun DE, Hao Y, Wang J, Wang C, Chen X. Cell-type-specific labeling and profiling of glycans in living mice. Nat Chem Biol 2022; 18:625-633. [PMID: 35513511 DOI: 10.1038/s41589-022-01016-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 03/15/2022] [Indexed: 11/09/2022]
Abstract
Metabolic labeling of glycans with clickable unnatural sugars has enabled glycan analysis in multicellular systems. However, cell-type-specific labeling of glycans in vivo remains challenging. Here we develop genetically encoded metabolic glycan labeling (GeMGL), a cell-type-specific strategy based on a bump-and-hole pair of an unnatural sugar and its matching engineered enzyme. N-pentynylacetylglucosamine (GlcNAl) serves as a bumped analog of N-acetylglucosamine (GlcNAc) that is specifically incorporated into glycans of cells expressing a UDP-GlcNAc pyrophosphorylase mutant, AGX2F383G. GeMGL with the 1,3-di-O-propionylated GlcNAl (1,3-Pr2GlcNAl) and AGX2F383G pair was demonstrated in cell cocultures, and used for specific labeling of glycans in mouse xenograft tumors. By generating a transgenic mouse line with AGX2F383G expressed under a cardiomyocyte-specific promoter, we performed specific imaging of cardiomyocyte glycans in the heart and identified 582 cardiomyocyte O-GlcNAcylated proteins with no interference from other cardiac cell types. GeMGL will facilitate cell-type-specific glycan imaging and glycoproteomics in various tissues and disease models.
Collapse
Affiliation(s)
- Xinqi Fan
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Qitao Song
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - De-En Sun
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Yi Hao
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Jingyang Wang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Chunting Wang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China. .,Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China. .,Synthetic and Functional Biomolecules Center, Peking University, Beijing, China. .,Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China.
| |
Collapse
|
36
|
Vang S, Cochran P, Sebastian Domingo J, Krick S, Barnes JW. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites 2022; 12:metabo12040316. [PMID: 35448503 PMCID: PMC9026683 DOI: 10.3390/metabo12040316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease of complex etiology. Cases of PAH that do not receive therapy after diagnosis have a low survival rate. Multiple reports have shown that idiopathic PAH, or IPAH, is associated with metabolic dysregulation including altered bioavailability of nitric oxide (NO) and dysregulated glucose metabolism. Multiple processes such as increased proliferation of pulmonary vascular cells, angiogenesis, apoptotic resistance, and vasoconstriction may be regulated by the metabolic changes demonstrated in PAH. Recent reports have underscored similarities between metabolic abnormalities in cancer and IPAH. In particular, increased glucose uptake and altered glucose utilization have been documented and have been linked to the aforementioned processes. We were the first to report a link between altered glucose metabolism and changes in glycosylation. Subsequent reports have highlighted similar findings, including a potential role for altered metabolism and aberrant glycosylation in IPAH pathogenesis. This review will detail research findings that demonstrate metabolic dysregulation in PAH with an emphasis on glycobiology. Furthermore, this report will illustrate the similarities in the pathobiology of PAH and cancer and highlight the novel findings that researchers have explored in the field.
Collapse
|
37
|
Lee K, Lee J, Lee P, Jeon BC, Song MY, Kwak S, Lee J, Kim J, Kim D, Kim JH, Tesh VL, Lee M, Park S. Inhibition of O-GlcNAcylation protects from Shiga toxin-mediated cell injury and lethality in host. EMBO Mol Med 2022; 14:e14678. [PMID: 34842355 PMCID: PMC8749473 DOI: 10.15252/emmm.202114678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/09/2022] Open
Abstract
Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli (EHEC) are the major virulence factors responsible for hemorrhagic colitis, which can lead to life-threatening systemic complications including acute renal failure (hemolytic uremic syndrome) and neuropathy. Here, we report that O-GlcNAcylation, a type of post-translational modification, was acutely increased upon induction of endoplasmic reticulum (ER) stress in host cells by Stxs. Suppression of the abnormal Stx-mediated increase in O-GlcNAcylation effectively inhibited apoptotic and inflammatory responses in Stx-susceptible cells. The protective effect of O-GlcNAc inhibition for Stx-mediated pathogenic responses was also verified using three-dimensional (3D)-cultured spheroids or organoids mimicking the human kidney. Treatment with an O-GlcNAcylation inhibitor remarkably improved the major disease symptoms and survival rate for mice intraperitoneally injected with a lethal dose of Stx. In conclusion, this study elucidates O-GlcNAcylation-dependent pathogenic mechanisms of Stxs and demonstrates that inhibition of aberrant O-GlcNAcylation is a potential approach to treat Stx-mediated diseases.
Collapse
Affiliation(s)
- Kyung‐Soo Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jieun Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Pureum Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Bong Chan Jeon
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
- Immunotherapy Convergence Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Min Yeong Song
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Sojung Kwak
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Jungwoon Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jun‐Seob Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheonKorea
| | - Doo‐Jin Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Ji Hyung Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and ImmunologyCollege of MedicineTexas A&M UniversityBryanTXUSA
| | - Moo‐Seung Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Sung‐Kyun Park
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| |
Collapse
|
38
|
Cai L. Prevention or therapy of the diabetic cardiomyopathy by fine O-GlcNAcylation balance: hopes and concerns. Cardiovasc Res 2022; 118:7-9. [PMID: 34254135 DOI: 10.1093/cvr/cvab226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Lu Cai
- Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
39
|
Ishikita A, Matsushima S, Ikeda S, Okabe K, Nishimura R, Tadokoro T, Enzan N, Yamamoto T, Sada M, Tsutsui Y, Miyake R, Ikeda M, Ide T, Kinugawa S, Tsutsui H. GFAT2 mediates cardiac hypertrophy through HBP-O-GlcNAcylation-Akt pathway. iScience 2021; 24:103517. [PMID: 34934932 DOI: 10.1016/j.isci.2021.103517] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 09/25/2021] [Accepted: 11/23/2021] [Indexed: 01/14/2023] Open
Abstract
Molecular mechanisms mediating cardiac hypertrophy by glucose metabolism are incompletely understood. Hexosamine biosynthesis pathway (HBP), an accessory pathway of glycolysis, is known to be involved in the attachment of O-linked N-acetylglucosamine motif (O-GlcNAcylation) to proteins, a post-translational modification. We here demonstrate that glutamine-fructose-6-phosphate amidotransferase 2 (GFAT2), a critical HBP enzyme, is a major isoform of GFAT in the heart and is increased in response to several hypertrophic stimuli, including isoproterenol (ISO). Knockdown of GFAT2 suppresses ISO-induced cardiomyocyte hypertrophy, accompanied by suppression of Akt O-GlcNAcylation and activation. Knockdown of GFAT2 does not affect anti-hypertrophic effect by Akt inhibition. Administration of glucosamine, a substrate of HBP, induces protein O-GlcNAcylation, Akt activation, and cardiomyocyte hypertrophy. In mice, 6-diazo-5-oxo-L-norleucine, an inhibitor of GFAT, attenuates ISO-induced protein O-GlcNAcylation, Akt activation, and cardiac hypertrophy. Our results demonstrate that GFAT2 mediates cardiomyocyte hypertrophy by HBP-O-GlcNAcylation-Akt pathway and could be a critical therapeutic target of cardiac hypertrophy.
Collapse
Affiliation(s)
- Akihito Ishikita
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kosuke Okabe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryohei Nishimura
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Nobuyuki Enzan
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taishi Yamamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Sada
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitomo Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Miyake
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomomi Ide
- Department of Experimental and Clinical Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
40
|
Loaeza-Reyes KJ, Zenteno E, Moreno-Rodríguez A, Torres-Rosas R, Argueta-Figueroa L, Salinas-Marín R, Castillo-Real LM, Pina-Canseco S, Cervera YP. An Overview of Glycosylation and its Impact on Cardiovascular Health and Disease. Front Mol Biosci 2021; 8:751637. [PMID: 34869586 PMCID: PMC8635159 DOI: 10.3389/fmolb.2021.751637] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022] Open
Abstract
The cardiovascular system is a complex and well-organized system in which glycosylation plays a vital role. The heart and vascular wall cells are constituted by an array of specific receptors; most of them are N- glycosylated and mucin-type O-glycosylated. There are also intracellular signaling pathways regulated by different post-translational modifications, including O-GlcNAcylation, which promote adequate responses to extracellular stimuli and signaling transduction. Herein, we provide an overview of N-glycosylation and O-glycosylation, including O-GlcNAcylation, and their role at different levels such as reception of signal, signal transduction, and exogenous molecules or agonists, which stimulate the heart and vascular wall cells with effects in different conditions, like the physiological status, ischemia/reperfusion, exercise, or during low-grade inflammation in diabetes and aging. Furthermore, mutations of glycosyltransferases and receptors are associated with development of cardiovascular diseases. The knowledge on glycosylation and its effects could be considered biochemical markers and might be useful as a therapeutic tool to control cardiovascular diseases.
Collapse
Affiliation(s)
- Karen Julissa Loaeza-Reyes
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico.,Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Rafael Torres-Rosas
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Liliana Argueta-Figueroa
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico.,Conacyt - Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Roberta Salinas-Marín
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Lizet Monserrat Castillo-Real
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Yobana Pérez Cervera
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico.,Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| |
Collapse
|
41
|
Zou L, Collins HE, Young ME, Zhang J, Wende AR, Darley-Usmar VM, Chatham JC. The Identification of a Novel Calcium-Dependent Link Between NAD + and Glucose Deprivation-Induced Increases in Protein O-GlcNAcylation and ER Stress. Front Mol Biosci 2021; 8:780865. [PMID: 34950703 PMCID: PMC8691773 DOI: 10.3389/fmolb.2021.780865] [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: 09/21/2021] [Accepted: 11/22/2021] [Indexed: 01/19/2023] Open
Abstract
The modification of proteins by O-linked β-N-acetylglucosamine (O-GlcNAc) is associated with the regulation of numerous cellular processes. Despite the importance of O-GlcNAc in mediating cellular function our understanding of the mechanisms that regulate O-GlcNAc levels is limited. One factor known to regulate protein O-GlcNAc levels is nutrient availability; however, the fact that nutrient deficient states such as ischemia increase O-GlcNAc levels suggests that other factors also contribute to regulating O-GlcNAc levels. We have previously reported that in unstressed cardiomyocytes exogenous NAD+ resulted in a time and dose dependent decrease in O-GlcNAc levels. Therefore, we postulated that NAD+ and cellular O-GlcNAc levels may be coordinately regulated. Using glucose deprivation as a model system in an immortalized human ventricular cell line, we examined the influence of extracellular NAD+ on cellular O-GlcNAc levels and ER stress in the presence and absence of glucose. We found that NAD+ completely blocked the increase in O-GlcNAc induced by glucose deprivation and suppressed the activation of ER stress. The NAD+ metabolite cyclic ADP-ribose (cADPR) had similar effects on O-GlcNAc and ER stress suggesting a common underlying mechanism. cADPR is a ryanodine receptor (RyR) agonist and like caffeine, which also activates the RyR, both mimicked the effects of NAD+. SERCA inhibition, which also reduces ER/SR Ca2+ levels had similar effects to both NAD+ and cADPR on O-GlcNAc and ER stress responses to glucose deprivation. The observation that NAD+, cADPR, and caffeine all attenuated the increase in O-GlcNAc and ER stress in response to glucose deprivation, suggests a potential common mechanism, linked to ER/SR Ca2+ levels, underlying their activation. Moreover, we showed that TRPM2, a plasma membrane cation channel was necessary for the cellular responses to glucose deprivation. Collectively, these findings support a novel Ca2+-dependent mechanism underlying glucose deprivation induced increase in O-GlcNAc and ER stress.
Collapse
Affiliation(s)
- Luyun Zou
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Helen E. Collins
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Martin E. Young
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianhua Zhang
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States,Birmingham VA Medical Center, Birmingham, AL, United States
| | - Adam R. Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Victor M. Darley-Usmar
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John C. Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: John C. Chatham,
| |
Collapse
|
42
|
Ataxin-10 Inhibits TNF- α-Induced Endothelial Inflammation via Suppressing Interferon Regulatory Factor-1. Mediators Inflamm 2021; 2021:7042148. [PMID: 34858081 PMCID: PMC8632433 DOI: 10.1155/2021/7042148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/01/2021] [Indexed: 02/08/2023] Open
Abstract
Endothelial inflammation is a crucial event in the initiation of atherosclerosis. Here, we identify Ataxin-10 protein as a novel negative modulator of endothelial activation by suppressing IRF-1 transcription activity. The protein level of Ataxin-10 is relatively higher in human vascular endothelial cells, which can be significantly suppressed by TNF-α in both HUVECs and HLMECs. Overexpression of Ataxin-10 markedly inhibited the mRNA expressions of VCAM-1 and several cytokines including MCP-1, CXCL-1, CCL-5, and TNF-α; thus, it can also suppress monocyte adhesion to endothelial cells. Accordingly, Ataxin-10 silencing promoted endothelial inflammation. However, Ataxin-10 did not affect the MAPK/NF-κB signaling pathway stimulated by TNF-α in HUVECs. Using the yeast two-hybrid assay, we found that Ataxin-10 can directly bind to interferon regulatory factor-1 (IRF-1). Upon TNF-α stimulation, Ataxin-10 promoted the cytoplasmic localization of IRF-1, which inhibited the transcription of VCAM-1. Moreover, knockdown of IRF-1 can eliminate the effect of Ataxin-10 on the expression of VCAM-1 in HUVECs induced by TNF-α. Taken together, these results indicate that Ataxin-10 inhibits endothelial cell activation and may serve as a promising therapeutic target for some vascular inflammatory-related diseases such as atherosclerosis.
Collapse
|
43
|
Writing and erasing O-GlcNAc from target proteins in cells. Biochem Soc Trans 2021; 49:2891-2901. [PMID: 34783346 DOI: 10.1042/bst20210865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
O-linked N-acetylglucosamine (O-GlcNAc) is a widespread reversible modification on nucleocytoplasmic proteins that plays an important role in many biochemical processes and is highly relevant to numerous human diseases. The O-GlcNAc modification has diverse functional impacts on individual proteins and glycosites, and methods for editing this modification on substrates are essential to decipher these functions. Herein, we review recent progress in developing methods for O-GlcNAc regulation, with a focus on methods for editing O-GlcNAc with protein- and site-selectivity in cells. The applications, advantages, and limitations of currently available strategies for writing and erasing O-GlcNAc and future directions are also discussed. These emerging approaches to manipulate O-GlcNAc on a target protein in cells will greatly accelerate the development of functional studies and enable therapeutic interventions in the O-GlcNAc field.
Collapse
|
44
|
Weng Y, Wang Z, Fukuhara Y, Tanai A, Ikegame M, Yamada D, Takarada T, Izawa T, Hayano S, Yoshida K, Kamioka H, Okamura H. O-GlcNAcylation drives calcium signaling toward osteoblast differentiation: A bioinformatics-oriented study. Biofactors 2021; 47:992-1015. [PMID: 34418170 DOI: 10.1002/biof.1774] [Citation(s) in RCA: 10] [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: 05/31/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
This study aimed to reveal the possible mechanisms by which O-linked-N-acetylglucosaminylation (O-GlcNAcylation) regulates osteoblast differentiation using a series of bioinformatics-oriented experiments. To examine the influence of O-GlcNAcylation levels on osteoblast differentiation, osteoblastic MC3T3-E1 cells were treated with O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) inhibitors. Correlations between the levels of O-GlcNAcylation and the expression of osteogenic markers as well as OGT were evaluated by qPCR and western blotting. The O-GlcNAcylated proteins assumed to correlate with Runx2 expression were retrieved from several public databases and used for further bioinformatics analysis. Following the findings of the bioinformatics analysis, intracellular calcium ([Ca2+ ]i ) was monitored in the cells treated with OGT and OGA inhibitors using a confocal laser-scanning microscope (CLS). The interaction effect between O-GlcNAcylation and [Ca2+ ]i on osteogenic marker expression was determined using stable OGT knockdown MC3T3-E1 cells. O-GlcNAcylation was positively associated with osteoblast differentiation. The time-course profile of global O-GlcNAcylated proteins showed a distinctive pattern with different molecular weights during osteoblast differentiation. The expression pattern of several O-GlcNAcylated proteins was significantly similar to that of Runx2 expression. Bioinformatic analysis of the retrieved Runx2-related-O-GlcNAcylated-proteins revealed the importance of [Ca2+ ]i . CLS showed that alteration of O-GlcNAcylation rapidly changed [Ca2+ ]i in MC3T3-E1 cells. O-GlcNAcylation and [Ca2+ ]i showed an interaction effect on the expression of osteogenic markers. OGT knockdown disrupted the [Ca2+ ]i -induced expression changes of osteogenic markers. O-GlcNAcylation interacts with [Ca2+ ]i and elicits osteoblast differentiation by regulating the expression of osteogenic markers.
Collapse
Affiliation(s)
- Yao Weng
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ziyi Wang
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoko Fukuhara
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Airi Tanai
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mika Ikegame
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takashi Izawa
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoru Hayano
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kaya Yoshida
- Department of Oral Healthcare Education, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hirohiko Okamura
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
45
|
Aung LHH, Jumbo JCC, Wang Y, Li P. Therapeutic potential and recent advances on targeting mitochondrial dynamics in cardiac hypertrophy: A concise review. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:416-443. [PMID: 34484866 PMCID: PMC8405900 DOI: 10.1016/j.omtn.2021.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pathological cardiac hypertrophy begins as an adaptive response to increased workload; however, sustained hemodynamic stress will lead it to maladaptation and eventually cardiac failure. Mitochondria, being the powerhouse of the cells, can regulate cardiac hypertrophy in both adaptive and maladaptive phases; they are dynamic organelles that can adjust their number, size, and shape through a process called mitochondrial dynamics. Recently, several studies indicate that promoting mitochondrial fusion along with preventing mitochondrial fission could improve cardiac function during cardiac hypertrophy and avert its progression toward heart failure. However, some studies also indicate that either hyperfusion or hypo-fission could induce apoptosis and cardiac dysfunction. In this review, we summarize the recent knowledge regarding the effects of mitochondrial dynamics on the development and progression of cardiac hypertrophy with particular emphasis on the regulatory role of mitochondrial dynamics proteins through the genetic, epigenetic, and post-translational mechanisms, followed by discussing the novel therapeutic strategies targeting mitochondrial dynamic pathways.
Collapse
Affiliation(s)
- Lynn Htet Htet Aung
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.,Center for Bioinformatics, Institute for Translational Medicine, School of Basic Science, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Juan Carlos Cueva Jumbo
- School of Preclinical Medicine, Nanobody Research Center, Guangxi Medical University, Nanning 530021, China
| | - Yin Wang
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.,Center for Bioinformatics, Institute for Translational Medicine, School of Basic Science, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
46
|
Guo X, Deng Y, Zhan L, Shang J, Liu H. O‑GlcNAcylation contributes to intermittent hypoxia‑associated vascular dysfunction via modulation of MAPKs but not CaMKII pathways. Mol Med Rep 2021; 24:744. [PMID: 34435655 PMCID: PMC8430318 DOI: 10.3892/mmr.2021.12384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/28/2021] [Indexed: 11/10/2022] Open
Abstract
Intermittent hypoxia (IH) leads to vascular dysfunction, and O-linked-β-N-acetylglucosamine (O-GlcNAc)ylation may regulate vascular reactivity through the modulation of intracellular signaling. The present study hypothesized that O-GlcNAc modifications contributed to the vascular effects of acute IH (AIH) and chronic IH (CIH) through the MAPK and Ca2+/calmodulin-dependent kinase II (CaMKII) pathways. Rat aortic and mesenteric segments were incubated with DMSO, O-GlcNAcase (OGA) or O-GlcNAc transferase (OGT) inhibitor under either normoxic or AIH conditions for 3 h, and arterial function was then assessed. Meanwhile, arteries isolated from control and CIH rats were exposed to 3 h of incubation under normoxic conditions using DMSO, OGA or OGT as an inhibitor, before assessing arterial reactivity. CIH was found to increase the expression of vascular O-GlcNAc protein and OGT, phosphorylate p38 MAPK and ERK1/2, and decrease OGA levels, but it had no effects on phosphorylated CaMKII levels. OGA inhibition increased global O-GlcNAcylation and the phosphorylation of p38 MAPK, ERK1/2 and CaMKII, whereas OGT blockade had the opposite effects. OGA inhibition preserved acetylcholine-induced relaxation in AIH arteries, whereas OGT blockade attenuated the relaxation responses of arteries under normoxic conditions or undergoing AIH treatments. However, the impairment of acetylcholine dilation in CIH mesenteric arteries was improved. CIH artery contraction was increased following angiotensin II (Ang II) exposure. Blockade of p38 MAPK and ERK1/2, but not CaMKII, attenuated Ang II-induced contractile responses in CIH arteries isolated from the non-OGT inhibitor-treated groups. OGT inhibition significantly blocked contractile responses to Ang II and abolished the inhibitory effects of MAPK inhibitors. These findings indicated that O-GlcNAcylation regulates IH-induced vascular dysfunction, at least partly by modulating MAPK, but not CaMKII, signaling pathways.
Collapse
Affiliation(s)
- Xueling Guo
- Department of Critical Care Medicine, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Yan Deng
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of The Ministry of Health, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Linghui Zhan
- Department of Critical Care Medicine, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Jin Shang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of The Ministry of Health, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of The Ministry of Health, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
47
|
Guttzeit S, Backs J. Post-translational modifications talk and crosstalk to class IIa histone deacetylases. J Mol Cell Cardiol 2021; 162:53-61. [PMID: 34416247 DOI: 10.1016/j.yjmcc.2021.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/08/2021] [Accepted: 08/12/2021] [Indexed: 12/30/2022]
Abstract
Epigenetic modifications, such as histone or DNA modifications are key regulators of gene transcription and changes are often associated with maladaptive processes underlying cardiovascular disease. Epigenetic regulators therefore likely play a crucial role in cardiomyocyte homeostasis and facilitate the cellular adaption to various internal and external stimuli, responding to different intercellular and extracellular cues. Class IIa histone deacetylases are a class of epigenetic regulators that possess a myriad of post-transcriptional modification sites that modulate their activity in response to oxidative stress, altered catecholamine signalling or changes in the cellular metabolism. This review summaries the known reversible, post-translational modifications (PTMs) of class IIa histone deacetylases (HDACs) that ultimately drive transcriptional changes in homeostasis and disease. We also highlight the idea of a crosstalk of various PTMs on class IIa HDACs potentially leading to compensatory or synergistic effects on the class IIa HDAC-regulated cell behavior.
Collapse
Affiliation(s)
- Sebastian Guttzeit
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
48
|
Karlstaedt A, Barrett M, Hu R, Gammons ST, Ky B. Cardio-Oncology: Understanding the Intersections Between Cardiac Metabolism and Cancer Biology. JACC Basic Transl Sci 2021; 6:705-718. [PMID: 34466757 PMCID: PMC8385559 DOI: 10.1016/j.jacbts.2021.05.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 12/24/2022]
Abstract
An important priority in the cardiovascular care of oncology patients is to reduce morbidity and mortality, and improve the quality of life in cancer survivors through cross-disciplinary efforts. The rate of survival in cancer patients has improved dramatically over the past decades. Nonetheless, survivors may be more likely to die from cardiovascular disease in the long term, secondary, not only to the potential toxicity of cancer therapeutics, but also to the biology of cancer. In this context, efforts from basic and translational studies are crucial to understanding the molecular mechanisms causal to cardiovascular disease in cancer patients and survivors, and identifying new therapeutic targets that may prevent and treat both diseases. This review aims to highlight our current understanding of the metabolic interaction between cancer and the heart, including potential therapeutic targets. An overview of imaging techniques that can support both research studies and clinical management is also provided. Finally, this review highlights opportunities and challenges that are necessary to advance our understanding of metabolism in the context of cardio-oncology.
Collapse
Key Words
- 99mTc-MIBI, 99mtechnetium-sestamibi
- CVD, cardiovascular disease
- D2-HG, D-2-hydroxyglutarate
- FAO, fatty acid oxidation
- FASN, fatty acid synthase
- GLS, glutaminase
- HF, heart failure
- IDH, isocitrate dehydrogenase
- IGF, insulin-like growth factor
- MCT1, monocarboxylate transporter 1
- MRS, magnetic resonance spectroscopy
- PDH, pyruvate dehydrogenase
- PET, positron emission tomography
- PI3K, insulin-activated phosphoinositide-3-kinase
- PTM, post-translational modification
- SGLT2, sodium glucose co-transporter 2
- TRF, time-restricted feeding
- [18F]FDG, 2-deoxy-2-[fluorine-18]fluoro-D-glucose
- cancer
- cardio-oncology
- heart failure
- metabolism
- oncometabolism
- α-KG, α-ketoglutarate
Collapse
Affiliation(s)
- Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Matthew Barrett
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ray Hu
- Departments of Medicine and Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seth Thomas Gammons
- Department of Cancer Systems Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Bonnie Ky
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Departments of Medicine and Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Zhu WZ, Ledee D, Olson AK. Temporal regulation of protein O-GlcNAc levels during pressure-overload cardiac hypertrophy. Physiol Rep 2021; 9:e14965. [PMID: 34337900 PMCID: PMC8326887 DOI: 10.14814/phy2.14965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/03/2022] Open
Abstract
Protein posttranslational modifications (PTMs) by O-linked β-N-acetylglucosamine (O-GlcNAc) rise during pressure-overload hypertrophy (POH) to affect hypertrophic growth. The hexosamine biosynthesis pathway (HBP) branches from glycolysis to make the moiety for O-GlcNAcylation. It is speculated that greater glucose utilization during POH augments HBP flux to increase O-GlcNAc levels; however, recent results suggest glucose availability does not primarily regulate cardiac O-GlcNAc levels. We hypothesize that induction of key enzymes augment protein O-GlcNAc levels primarily during active myocardial hypertrophic growth and remodeling with early pressure overload. We further speculate that downregulation of protein O-GlcNAcylation inhibits ongoing hypertrophic growth during prolonged pressure overload with established hypertrophy. We used transverse aortic constriction (TAC) to create POH in C57/Bl6 mice. Experimental groups were sham, 1-week TAC (1wTAC) for early hypertrophy, or 6-week TAC (6wTAC) for established hypertrophy. We used western blots to determine O-GlcNAc regulation. To assess the effect of increased protein O-GlcNAcylation with established hypertrophy, mice received thiamet-g (TG) starting 4 weeks after TAC. Protein O-GlcNAc levels were significantly elevated in 1wTAC versus Sham with a fall in 6wTAC. OGA, which removes O-GlcNAc from proteins, fell in 1wTAC versus sham. GFAT is the rate-limiting HBP enzyme and the isoform GFAT1 substantially rose in 1wTAC. With established hypertrophy, TG increased protein O-GlcNAc levels but did not affect cardiac mass. In summary, protein O-GlcNAc levels vary during POH with elevations occurring during active hypertrophic growth early after TAC. O-GlcNAc levels appear to be regulated by changes in key enzyme levels. Increasing O-GlcNAc levels during established hypertrophy did not restart hypertrophic growth.
Collapse
Affiliation(s)
| | - Dolena Ledee
- Seattle Children’s Research InstituteSeattleWAUSA
- Division of CardiologyDepartment of PediatricsUniversity of WashingtonSeattleWAUSA
| | - Aaron K. Olson
- Seattle Children’s Research InstituteSeattleWAUSA
- Division of CardiologyDepartment of PediatricsUniversity of WashingtonSeattleWAUSA
| |
Collapse
|
50
|
Ou W, Liang Y, Qin Y, Wu W, Xie M, Zhang Y, Zhang Y, Ji L, Yu H, Li T. Hypoxic acclimation improves cardiac redox homeostasis and protects heart against ischemia-reperfusion injury through upregulation of O-GlcNAcylation. Redox Biol 2021; 43:101994. [PMID: 33964586 PMCID: PMC8121980 DOI: 10.1016/j.redox.2021.101994] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/11/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023] Open
Abstract
Ischemia-reperfusion (I/R) injury is detrimental to cardiovascular system. Alteration in glucose metabolism has been recognized as an important adaptive response under hypoxic conditions. However, the biological benefits underlying this metabolic phenotype remain to be elucidated. This study was designed to investigate the impact of hypoxic acclimation (HA) on cardiac I/R injury and the antioxidative mechanism(s). Male adult mice were acclimated in a hypoxic chamber (10% oxygen [O2]) for 8 h/day for 14 days, and then subjected to cardiac I/R injury by ligation of left anterior descending coronary artery for 30 min and reperfusion for 24 h or 7 days. Our results showed that HA attenuated oxidative stress and reduced infarct size in the I/R hearts. This cardioprotective effect is coupled with an elevation of protein O-linked N-acetylglucosamine (O-GlcNAc) modification partially due to inflammatory stimulation. Hyperglycosylation activated glucose-6-phosphate dehydrogenase (G6PDH), the rate-limiting enzyme in the pentose phosphate pathway, resulting in an upregulation of NADPH/NADP+ and GSH/GSSG couples and enhancement of redox homeostasis in the heart. Pharmacological suppression of O-GlcNAcylation totally abolished the influence of HA on the G6PDH activity, redox balance and post-I/R damage in the hearts and cultured cardiomyocytes, whereby augmentation of O-GlcNAcylation further enhanced the benefits, suggesting a central role of O-GlcNAcylation in HA-initiated antioxidative and cardioprotective effects. These findings, therefore, identified HA as a promising anti-I/R strategy for the heart and proposed O-GlcNAc modification of G6PDH as a therapeutic target in ischemic heart disease.
Collapse
Affiliation(s)
- Wei Ou
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China; Department of Anesthesiology, Nanchong Central Hospital, Nanchong, 637000, China
| | - Yu Liang
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yu Qin
- Department of Anesthesiology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wei Wu
- Department of Anesthesiology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Maodi Xie
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yabing Zhang
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yarong Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Liwei Ji
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|