1
|
He N, Li Y, Liu F, Dong X, Ma D. Adipocytes regulate monocyte development through the OGT-NEFA-CD36/FABP4 pathway in high-fat diet-induced obesity. Cell Death Dis 2025; 16:401. [PMID: 40389445 DOI: 10.1038/s41419-025-07721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/17/2025] [Accepted: 05/06/2025] [Indexed: 05/21/2025]
Abstract
Obesity, resulting from excessive adipocyte accumulation, is a primary risk for various diseases. Although its impact on hematopoietic stem cell (HSC) function has been reported, its effects on HSC differentiation remain controversial. O-GlcNAc transferase (OGT), which catalyzes the attachment of N-acetylglucosamine to serine and threonine residues in proteins, acts as a metabolic sensor capable of regulating diverse physiological processes. This study demonstrates that obesity is associated with higher peripheral monocyte levels. Adipocyte OGT is crucial for monocyte development in high-fat diet (HFD)-induced obesity, promoting an increase in peripheral blood monocytes through transcriptional activation of nonesterified fatty acids (NEFA), a critical energy substrate. Loss of adipocyte OGT decreases serum NEFA levels, reduces white adipose tissue, and inhibits HSC differentiation into monocytes in HFD-induced obesity. Mechanistically, the regulated effect of adipocyte OGT on monocyte development may be mediated by NEFA-cluster of differentiation 36/fatty acid binding protein 4 (CD36/FABP4) pathway in HSCs in HFD-induced obesity. These findings establish the critical role of adipocyte OGT in hematopoietic homeostasis and monocyte development.
Collapse
Affiliation(s)
- Na He
- Advanced Medical Research InstiTabletute, Shandong University, Shandong, 250012, China
- Department of Hematology, Qilu Hospital of Shandong University, Shandong, 250012, China
| | - Yingjie Li
- Department of Health Management Center, Qilu Hospital of Shandong University, Shandong, 250012, China
| | - Fabao Liu
- Advanced Medical Research InstiTabletute, Shandong University, Shandong, 250012, China
| | - Xifeng Dong
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital of Shandong University, Shandong, 250012, 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
|
Mycroft-West CJ, Leanca MA, Wu L. Structural glycobiology - from enzymes to organelles. Biochem Soc Trans 2025; 53:BST20241119. [PMID: 39889286 DOI: 10.1042/bst20241119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 02/02/2025]
Abstract
Biological carbohydrate polymers represent some of the most complex molecules in life, enabling their participation in a huge range of physiological functions. The complexity of biological carbohydrates arises from an extensive enzymatic repertoire involved in their construction, deconstruction and modification. Over the past decades, structural studies of carbohydrate processing enzymes have driven major insights into their mechanisms, supporting associated applications across medicine and biotechnology. Despite these successes, our understanding of how multienzyme networks function to create complex polysaccharides is still limited. Emerging techniques such as super-resolution microscopy and cryo-electron tomography are now enabling the investigation of native biological systems at near molecular resolutions. Here, we review insights from classical in vitro studies of carbohydrate processing, alongside recent in situ studies of glycosylation-related processes. While considerable technical challenges remain, the integration of molecular mechanisms with true biological context promises to transform our understanding of carbohydrate regulation, shining light upon the processes driving functional complexity in these essential biomolecules.
Collapse
Affiliation(s)
| | - Miron A Leanca
- The Rosalind Franklin Institute, Harwell Science & Innovation Campus, OX11 0QX, Didcot, UK
| | - Liang Wu
- The Rosalind Franklin Institute, Harwell Science & Innovation Campus, OX11 0QX, Didcot, UK
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, OX3 7BN, Oxford, UK
| |
Collapse
|
4
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
5
|
Crisóstomo T, Luzes R, Gonçalves MLL, Pardal MAE, Muzi-Filho H, Costa-Sarmento G, Mello DB, Vieyra A. Male Wistar Rats Chronically Fed with a High-Fat Diet Develop Inflammatory and Ionic Transport Angiotensin-(3-4)-Sensitive Myocardial Lesions but Preserve Echocardiographic Parameters. Int J Mol Sci 2024; 25:12474. [PMID: 39596537 PMCID: PMC11594684 DOI: 10.3390/ijms252212474] [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: 07/12/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
The central aim of this study was to investigate whether male Wistar rats chronically fed a high-fat diet (HFD) over 106 days present high levels of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), and Na+ and Ca2+ transport alterations in the left ventricle, together with dyslipidemia and decreased glucose tolerance, and to investigate the influence of Ang-(3-4). The rats became moderately overweight with an expansion of visceral adiposity. Na+-transporting ATPases, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA2a), and the abundance of Angiotensin II receptors were studied together with lipid and glycemic profiles from plasma and left-ventricle echocardiographic parameters fractional shortening (FS) and ejection fraction (EF). IL-6 and TNF-α increased (62% and 53%, respectively), but returned to normal levels with Angiotensin-(3-4) administration after 106 days. Significant lipidogram alterations accompanied a decrease in glucose tolerance. Angiotensin II receptors abundance did not change. (Na+ + K+)ATPase and ouabain-resistant Na+-ATPase were downregulated and upregulated, respectively, but returned to normal values upon Angiotensin-(3-4) administration. SERCA2a lost its ability to respond to excess ATP. Echocardiography showed no changes in FS or EF. We conclude that being overweight causes an increase in Ang-(3-4)-sensitive IL-6 and TNF-α levels, and ion transport alterations in the left ventricle that could evolve into future heart dysfunction.
Collapse
Affiliation(s)
- Thuany Crisóstomo
- Leopoldo de Meis Institute of Medical Biochemistry, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Rafael Luzes
- Graduate Program in Translational Biomedicine (BIOTRANS), Grande Rio University (UNIGRANRIO), Duque de Caxias 25071-202, Brazil;
| | | | - Marco Antônio Estrela Pardal
- Carlos Chagas Filho Institute of Biophysics, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.A.E.P.); (H.M.-F.); (G.C.-S.)
| | - Humberto Muzi-Filho
- Carlos Chagas Filho Institute of Biophysics, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.A.E.P.); (H.M.-F.); (G.C.-S.)
| | - Glória Costa-Sarmento
- Carlos Chagas Filho Institute of Biophysics, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.A.E.P.); (H.M.-F.); (G.C.-S.)
| | - Debora B. Mello
- National Center for Structural Biology and Bioimaging/CENABIO, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Adalberto Vieyra
- Graduate Program in Translational Biomedicine (BIOTRANS), Grande Rio University (UNIGRANRIO), Duque de Caxias 25071-202, Brazil;
- Carlos Chagas Filho Institute of Biophysics, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.A.E.P.); (H.M.-F.); (G.C.-S.)
- National Center for Structural Biology and Bioimaging/CENABIO, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
6
|
Lei Y, Liu Q, Chen B, Wu F, Li Y, Dong X, Ma N, Wu Z, Zhu Y, Wang L, Fu Y, Liu Y, Song Y, Du M, Zhang H, Zhu J, Lyons TJ, Wang T, Hu J, Xu H, Chen M, Yan H, Wang X. Protein O-GlcNAcylation coupled to Hippo signaling drives vascular dysfunction in diabetic retinopathy. Nat Commun 2024; 15:9334. [PMID: 39472558 PMCID: PMC11522279 DOI: 10.1038/s41467-024-53601-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Metabolic disorder significantly contributes to diabetic vascular complications, including diabetic retinopathy, the leading cause of blindness in the working-age population. However, the molecular mechanisms by which disturbed metabolic homeostasis causes vascular dysfunction in diabetic retinopathy remain unclear. O-GlcNAcylation modification acts as a nutrient sensor particularly sensitive to ambient glucose. Here, we observe pronounced O-GlcNAc elevation in retina endothelial cells of diabetic retinopathy patients and mouse models. Endothelial-specific depletion or pharmacological inhibition of O-GlcNAc transferase effectively mitigates vascular dysfunction. Mechanistically, we find that Yes-associated protein (YAP) and Transcriptional co-activator with PDZ-binding motif (TAZ), key effectors of the Hippo pathway, are O-GlcNAcylated in diabetic retinopathy. We identify threonine 383 as an O-GlcNAc site on YAP, which inhibits its phosphorylation at serine 397, leading to its stabilization and activation, thereby promoting vascular dysfunction by inducing a pro-angiogenic and glucose metabolic transcriptional program. This work emphasizes the critical role of the O-GlcNAc-Hippo axis in the pathogenesis of diabetic retinopathy and suggests its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yi Lei
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiangyun Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Binggui Chen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fangfang Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yiming Li
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Nina Ma
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ziru Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuxin Fu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuming Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Yinting Song
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Mei Du
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Heng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jidong Zhu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Timothy J Lyons
- Division of Endocrinology, Diabetes and Metabolic Diseases at the Medical University of South Carolina, Charleston, SC, USA
| | - Ting Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junhao Hu
- Laboratory of Vascular Biology and Organ Homeostasis, Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Hua Yan
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medicine, Nankai University, Tianjin, China.
| | - Xiaohong Wang
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
7
|
Zhang L, Deng T, Pan S, Zhang M, Zhang Y, Yang C, Yang X, Tian G, Mi J. DeepO-GlcNAc: a web server for prediction of protein O-GlcNAcylation sites using deep learning combined with attention mechanism. Front Cell Dev Biol 2024; 12:1456728. [PMID: 39450274 PMCID: PMC11500328 DOI: 10.3389/fcell.2024.1456728] [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: 06/29/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Protein O-GlcNAcylation is a dynamic post-translational modification involved in major cellular processes and associated with many human diseases. Bioinformatic prediction of O-GlcNAc sites before experimental validation is a challenge task in O-GlcNAc research. Recent advancements in deep learning algorithms and the availability of O-GlcNAc proteomics data present an opportunity to improve O-GlcNAc site prediction. Objectives This study aims to develop a deep learning-based tool to improve O-GlcNAcylation site prediction. Methods We construct an annotated unbalanced O-GlcNAcylation data set and propose a new deep learning framework, DeepO-GlcNAc, using Long Short-Term Memory (LSTM), Convolutional Neural Networks (CNN) combined with attention mechanism. Results The ablation study confirms that the additional model components in DeepO-GlcNAc, such as attention mechanisms and LSTM, contribute positively to improving prediction performance. Our model demonstrates strong robustness across five cross-species datasets, excluding humans. We also compare our model with three external predictors using an independent dataset. Our results demonstrated that DeepO-GlcNAc outperforms the external predictors, achieving an accuracy of 92%, an average precision of 72%, a MCC of 0.60, and an AUC of 92% in ROC analysis. Moreover, we have implemented DeepO-GlcNAc as a web server to facilitate further investigation and usage by the scientific community. Conclusion Our work demonstrates the feasibility of utilizing deep learning for O-GlcNAc site prediction and provides a novel tool for O-GlcNAc investigation.
Collapse
Affiliation(s)
- Liyuan Zhang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Tingzhi Deng
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuijing Pan
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Minghui Zhang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Yusen Zhang
- School of Mathematics and Statistics, Shandong University, Weihai, Shandong, China
| | - Chunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaoyong Yang
- Department of Comparative Medicine, Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
8
|
Hu Y, Zou W, Zhang L, Zhang S, Hu L, Song Z, Kong S, Gao Y, Zhang J, Yang Y, Zheng J. TRPV3 facilitates lipolysis and attenuates diet-induced obesity via activation of the NRF2/FSP1 signaling axis. Free Radic Biol Med 2024; 221:155-168. [PMID: 38777204 DOI: 10.1016/j.freeradbiomed.2024.05.035] [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: 02/20/2024] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Transient receptor potential vanilloid (TRPV) ion channels play a crucial role in various cellular functions by regulating intracellular Ca2+ levels and have been extensively studied in the context of several metabolic diseases. However, the regulatory effects of TRPV3 in obesity and lipolysis are not well understood. In this study, utilizing a TRPV3 gain-of-function mouse model (TRPV3G568V/G568V), we assessed the metabolic phenotype of both TRPV3G568V/G568V mice and their control littermates, which were randomly assigned to either a 12-week high-fat diet or a control diet. We investigated the potential mechanisms underlying the role of TRPV3 in restraining obesity and promoting lipolysis both in vivo and in vitro. Our findings indicate that a high-fat diet led to significant obesity, characterized by increased epididymal and inguinal white adipose tissue weight and higher fat mass. However, the gain-of-function mutation in TRPV3 appeared to counteract these adverse effects by enhancing lipolysis in visceral fat through the upregulation of the major lipolytic enzyme, adipocyte triglyceride lipase (ATGL). In vitro experiments using carvacrol, a TRPV3 agonist, demonstrated the promotion of lipolysis and antioxidation in 3T3-L1 adipocytes after TRPV3 activation. Notably, carvacrol failed to stimulate Ca2+ influx, lipolysis, and antioxidation in 3T3-L1 adipocytes treated with BAPTA-AM, a cell-permeable calcium chelator. Our results revealed that TRPV3 activation induced the action of transcriptional factor nuclear factor erythroid 2-related factor 2 (NRF2), resulting in increased expression of ferroptosis suppressor protein 1 (FSP1) and superoxide dismutase2 (SOD2). Moreover, the inhibition of NRF2 impeded carvacrol-induced lipolysis and antioxidation in 3T3-L1 adipocytes, with downregulation of ATGL, FSP1, and SOD2. In summary, our study suggests that TRPV3 promotes visceral fat lipolysis and inhibits diet-induced obesity through the activation of the NRF2/FSP1 signaling axis. We propose that TRPV3 may be a potential therapeutic target in the treatment of obesity.
Collapse
Affiliation(s)
- Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing, China
| | - Wenyu Zou
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Shixuan Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Linghan Hu
- Genetic Skin Disease Center, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Zhongya Song
- Genetic Skin Disease Center, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Shenshen Kong
- Laboratory Animal Facility, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yong Yang
- Genetic Skin Disease Center, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
9
|
He M, Zhou X, Wang X. Glycosylation: mechanisms, biological functions and clinical implications. Signal Transduct Target Ther 2024; 9:194. [PMID: 39098853 PMCID: PMC11298558 DOI: 10.1038/s41392-024-01886-1] [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: 10/21/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Protein post-translational modification (PTM) is a covalent process that occurs in proteins during or after translation through the addition or removal of one or more functional groups, and has a profound effect on protein function. Glycosylation is one of the most common PTMs, in which polysaccharides are transferred to specific amino acid residues in proteins by glycosyltransferases. A growing body of evidence suggests that glycosylation is essential for the unfolding of various functional activities in organisms, such as playing a key role in the regulation of protein function, cell adhesion and immune escape. Aberrant glycosylation is also closely associated with the development of various diseases. Abnormal glycosylation patterns are closely linked to the emergence of various health conditions, including cancer, inflammation, autoimmune disorders, and several other diseases. However, the underlying composition and structure of the glycosylated residues have not been determined. It is imperative to fully understand the internal structure and differential expression of glycosylation, and to incorporate advanced detection technologies to keep the knowledge advancing. Investigations on the clinical applications of glycosylation focused on sensitive and promising biomarkers, development of more effective small molecule targeted drugs and emerging vaccines. These studies provide a new area for novel therapeutic strategies based on glycosylation.
Collapse
Affiliation(s)
- Mengyuan He
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
10
|
Su X, Patel N, Chen J, Chen Y, Zhou X, Mo X, Zhu S. Association between serum vitamin C and body mass index in adolescents aged 12-19 years. BMC Public Health 2024; 24:2067. [PMID: 39085802 PMCID: PMC11293171 DOI: 10.1186/s12889-024-19588-2] [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: 05/29/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Evidence on the association between serum vitamin C (sVC) levels and obesity is limited. This study aimed to explore the relationship between sVC and body mass index (BMI) in adolescents aged 12 to 19 years. METHODS We analyzed data from the National Health and Nutrition Examination Survey (NHANES) 2003-2006, with 3952 participants. sVC and BMI were independent variables and dependent variables, respectively. The associations of sVC with BMI were examined using multivariable linear regression models. Age, sex, and race/ethnicity were analyzed as subgroups. Then, we devised smooth curve fittings and saturation threshold analysis to address the nonlinear relationship. RESULTS sVC had a negative correlation with BMI after adjusting for all covariates (β: -1.020, 95% CI: -1.359, -0.680). In the subgroup analysis by age, sex, and race/ethnicity, there was still a negative correlation between sVC and BMI (p < 0.05). The analysis of saturation effects of sVC and BMI showed the relationship between sVC and BMI in female adolescents followed an N-shaped curve, whereas the relationship between sVC and BMI in adolescents aged 12-15 years and Mexican Americans followed a U-shaped curve. CONCLUSION Based on the results, proper vitamin C supplementation may be beneficial to weight loss. However, considering the threshold effect, large-scale and good-quality randomized controlled trials are required to obtain the optimal vitamin C level for weight control.
Collapse
Affiliation(s)
- Xiaoqi Su
- Department of Ultrasound, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- School of Public Health, Nanjing Medical University, Nanjing, 211666, China
| | - Jun Chen
- Department of Ultrasound, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ye Chen
- Department of Ultrasound, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xin Zhou
- Department of Ultrasound, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Shanliang Zhu
- Department of Ultrasound, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
11
|
Ohgaku S, Ida S, Ohashi N, Morino K, Ishikado A, Yanagimachi T, Murata K, Sato D, Ugi S, Nasiri A, Shulman GI, Maegawa H, Kume S, Fujita Y. O-GlcNAc modification in endothelial cells modulates adiposity via fat absorption from the intestine in mice. Heliyon 2024; 10:e34490. [PMID: 39130439 PMCID: PMC11315187 DOI: 10.1016/j.heliyon.2024.e34490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Endothelial cells have a crucial function in transporting and exchanging various nutrients. O-GlcNAcylation, mediated by O-GlcNAc transferase (OGT), involves the addition of N-acetylglucosamine to proteins and serves as an intracellular nutrient sensing mechanism. However, the role of O-GlcNAcylation in endothelial cells remains poorly understood. Objective This study investigated the role of O-GlcNAcylation in endothelial cells. Methods Endothelial-cell-specific Ogt -knockout mice (Ogt-ECKO) were generated by crossing Ogt-floxed mice (Ogt-flox) with VE-Cadherin Cre ERT2 mice. Ogt-ECKO mice and Ogt-flox control mice were subjected to a normal or high-fat diet, and their body weight, glucose metabolism, and lipid metabolism were examined. Results Ogt-ECKO mice exhibited reduced body weight compared with Ogt-flox control mice under a high-fat diet. Lipid absorption was significantly impaired in Ogt-ECKO mice. Changes in the intercellular junctions of small intestinal lacteal endothelial cells from a button-like to a zipper-like configuration were observed. Furthermore, Ogt-ECKO mice showed decreased expression of VEGFR3. The administration of a nitric oxide donor restored lipid absorption and reversed the morphological alterations in Ogt-ECKO mice. Conclusions These findings demonstrate the critical role of O-GlcNAcylation in endothelial cells in lipid absorption in the intestine through the modulation of lacteal junction morphology. These results provide novel insight into the metabolic regulatory mechanisms under physiological conditions and have implications for the development of new therapeutic strategies for obesity.
Collapse
Affiliation(s)
- Seiichiro Ohgaku
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Shogo Ida
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Natsuko Ohashi
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Katsutaro Morino
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
- Institutional Research Office, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Atsushi Ishikado
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
- R&D Department, Sunstar Inc., Osaka 569-1195, Japan
| | - Tsuyoshi Yanagimachi
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Koichiro Murata
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Daisuke Sato
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Satoshi Ugi
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Ali Nasiri
- Department of Medicine (Endocrinology), Yale School of Medicine, New Haven, CT 06520, USA
| | - Gerald I. Shulman
- Department of Medicine (Endocrinology), Yale School of Medicine, New Haven, CT 06520, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Yukihiro Fujita
- Department of Medicine, Shiga University of Medical Science, Otsu 520-2192, Japan
| |
Collapse
|
12
|
Persello A, Dupas T, Vergnaud A, Blangy-Letheule A, Aillerie V, Erraud A, Guilloux Y, Denis M, Lauzier B. Changes in transcriptomic landscape with macronutrients intake switch are independent from O-GlcNAcylation levels in heart throughout postnatal development in rats. Heliyon 2024; 10:e30526. [PMID: 38737268 PMCID: PMC11087977 DOI: 10.1016/j.heliyon.2024.e30526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/14/2024] Open
Abstract
Background Dietary intake and metabolism variations are associated with molecular changes and more particularly in the transcriptome. O-GlcNAcylation is a post-translational modification added and removed respectively by OGT and OGA. The UDP-GlcNAc, the substrate of OGT, is produced by UAP1 and UAP1L1. O-GlcNAcylation is qualified as a metabolic sensor and is involved in the modulation of gene expression. We wanted to unveil if O-GlcNAcylation is linking metabolic transition to transcriptomic changes and to highlight modifications of O-GlcNAcylation during the postnatal cardiac development. Methods Hearts were harvested from rats at birth (D0), before (D12) and after suckling to weaning transition with normal (D28) or delayed weaning diet from D12 to D28 (D28F). O-GlcNAcylation levels and proteins expression were evaluated by Western blot. Cardiac transcriptomes were evaluated via 3'SRP analysis. Results Cardiac O-GlcNAcylation levels and nucleocytoplasmic OGT (ncOGT) were decreased at D28 while full length OGA (OGA) was increased. O-GlcNAcylation levels did not changed with delayed weaning diet while ncOGT and OGA were respectively increased and decreased. Uapl1 was the only O-GlcNAcylation-related gene identified as differentially expressed throughout postnatal development. Conclusion Macronutrients switch promotes changes in the transcriptome landscape that are independent from O-GlcNAcylation levels. UAP1 and UAP1L1 are not the main regulator element of O-GlcNAcylation throughout postnatal development.
Collapse
Affiliation(s)
- Antoine Persello
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Thomas Dupas
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Amandine Vergnaud
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | | | - Virginie Aillerie
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Angélique Erraud
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Yannick Guilloux
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, CRCI2NA, F-44000, Nantes, France
| | - Manon Denis
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Benjamin Lauzier
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| |
Collapse
|
13
|
Peng Z, Zeng Y, Tan Q, He Q, Wang S, Wang J. 6-Gingerol alleviates ectopic lipid deposition in skeletal muscle by regulating CD36 translocation and mitochondrial function. Biochem Biophys Res Commun 2024; 708:149786. [PMID: 38493545 DOI: 10.1016/j.bbrc.2024.149786] [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: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Ectopic lipid deposition (ELD) and mitochondrial dysfunction are common causes of metabolic disorders in humans. Consuming too much fructose can result in mitochondrial dysfunction and metabolic disorders. 6-Gingerol, the main component of ginger (Zingiber officinale Roscoe), has been proven to alleviate metabolic disorders. This study seeks to examine the effects of 6-gingerol on metabolic disorders caused by fructose and uncover the underlying molecular mechanisms. In this study, the results showed that 6-Gingerol ameliorated high-fructose-induced metabolic disorders. Moreover, it inhibited CD36 membrane translocation, increased CD36 expression in the mitochondria, and decreased the O-GlcNAc modification of CD36 and OGT expression in vitro and vivo. In addition, 6-Gingerol enhanced the performance of mitochondria in the skeletal muscle and boosted the respiratory capability of L6 myotubes. This study provides a theoretical basis and new insights for the development of lipid-lowering drugs in clinical practice.
Collapse
Affiliation(s)
- Ze Peng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Yan Zeng
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Qi Tan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Qifeng He
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China.
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China; Chongqing College of Traditional Chinese Medicine, Chongqing, China.
| |
Collapse
|
14
|
Zhang X, Wang Q, Wang Y, Ma C, Zhao Q, Yin H, Li L, Wang D, Huang Y, Zhao Y, Shi X, Li X, Huang C. Interleukin-6 promotes visceral adipose tissue accumulation during aging via inhibiting fat lipolysis. Int Immunopharmacol 2024; 132:111906. [PMID: 38593501 DOI: 10.1016/j.intimp.2024.111906] [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: 11/03/2023] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Age-related visceral obesity could contribute to the development of cardiometabolic complications. The pathogenesis of visceral fat mass accumulation during the aging process remains complex and largely unknown. Interleukin-6 (IL-6) has emerged as one of the prominent inflammaging markers which are elevated in circulation during aging. However, the precise role of IL-6 in regulating age-related visceral adipose tissue accumulation remains uncertain. RESULTS A cross-sectional study including 77 older adults (≥65 years of age) was initially conducted. There was a significant positive association between serum IL-6 levels and visceral fat mass. We subsequently validated a modest but significant elevation in serum IL-6 levels in aged mice. Furthermore, we demonstrated that compared to wildtype control, IL-6 deficiency (IL-6 KO) significantly attenuated the accumulation of visceral adipose tissue during aging. Further metabolic characterization suggested that IL-6 deficiency resulted in improved lipid metabolism parameters and energy expenditure in aged mice. Moreover, histological examinations of adipose depots revealed that the absence of IL-6 ameliorated adipocyte hypertrophy in visceral adipose tissue of aged mice. Mechanically, the ablation of IL-6 could promote the PKA-mediated lipolysis and consequently mitigate lipid accumulation in adipose tissue in aged mice. CONCLUSION Our findings identify a detrimental role of IL-6 during the aging process by promoting visceral adipose tissue accumulation through inhibition of lipolysis. Therefore, strategies aimed at preventing or reducing IL-6 levels may potentially ameliorate age-related obesity and improve metabolism during aging.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Qingxuan Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yaru Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Chen Ma
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Qing Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Hongyan Yin
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Long Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Dongmei Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China; Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China
| | - Yinxiang Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yan Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xiulin Shi
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xuejun Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China.
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China.
| |
Collapse
|
15
|
Griseti E, Bello AA, Bieth E, Sabbagh B, Iacovoni JS, Bigay J, Laurell H, Čopič A. Molecular mechanisms of perilipin protein function in lipid droplet metabolism. FEBS Lett 2024; 598:1170-1198. [PMID: 38140813 DOI: 10.1002/1873-3468.14792] [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: 10/21/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Perilipins are abundant lipid droplet (LD) proteins present in all metazoans and also in Amoebozoa and fungi. Humans express five perilipins, which share a similar domain organization: an amino-terminal PAT domain and an 11-mer repeat region, which can fold into amphipathic helices that interact with LDs, followed by a structured carboxy-terminal domain. Variations of this organization that arose during vertebrate evolution allow for functional specialization between perilipins in relation to the metabolic needs of different tissues. We discuss how different features of perilipins influence their interaction with LDs and their cellular targeting. PLIN1 and PLIN5 play a direct role in lipolysis by regulating the recruitment of lipases to LDs and LD interaction with mitochondria. Other perilipins, particularly PLIN2, appear to protect LDs from lipolysis, but the molecular mechanism is not clear. PLIN4 stands out with its long repetitive region, whereas PLIN3 is most widely expressed and is used as a nascent LD marker. Finally, we discuss the genetic variability in perilipins in connection with metabolic disease, prominent for PLIN1 and PLIN4, underlying the importance of understanding the molecular function of perilipins.
Collapse
Affiliation(s)
- Elena Griseti
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Abdoul Akim Bello
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Eric Bieth
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
- Departement de Génétique Médicale, Centre Hospitalier Universitaire de Toulouse, France
| | - Bayane Sabbagh
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| | - Jason S Iacovoni
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Joëlle Bigay
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Alenka Čopič
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| |
Collapse
|
16
|
Sharma C, Hamza A, Boyle E, Donu D, Cen Y. Post-Translational Modifications and Diabetes. Biomolecules 2024; 14:310. [PMID: 38540730 PMCID: PMC10968569 DOI: 10.3390/biom14030310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetes and its associated complications have increasingly become major challenges for global healthcare. The current therapeutic strategies involve insulin replacement therapy for type 1 diabetes (T1D) and small-molecule drugs for type 2 diabetes (T2D). Despite these advances, the complex nature of diabetes necessitates innovative clinical interventions for effective treatment and complication prevention. Accumulative evidence suggests that protein post-translational modifications (PTMs), including glycosylation, phosphorylation, acetylation, and SUMOylation, play important roles in diabetes and its pathological consequences. Therefore, the investigation of these PTMs not only sheds important light on the mechanistic regulation of diabetes but also opens new avenues for targeted therapies. Here, we offer a comprehensive overview of the role of several PTMs in diabetes, focusing on the most recent advances in understanding their functions and regulatory mechanisms. Additionally, we summarize the pharmacological interventions targeting PTMs that have advanced into clinical trials for the treatment of diabetes. Current challenges and future perspectives are also provided.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Abu Hamza
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Emily Boyle
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Dickson Donu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
17
|
Hu YJ, Zhang X, Lv HM, Liu Y, Li SZ. Protein O-GlcNAcylation: The sweet hub in liver metabolic flexibility from a (patho)physiological perspective. Liver Int 2024; 44:293-315. [PMID: 38110988 DOI: 10.1111/liv.15812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023]
Abstract
O-GlcNAcylation is a dynamic, reversible and atypical O-glycosylation that regulates various cellular physiological processes via conformation, stabilisation, localisation, chaperone interaction or activity of target proteins. The O-GlcNAcylation cycle is precisely controlled by collaboration between O-GlcNAc transferase and O-GlcNAcase. Uridine-diphosphate-N-acetylglucosamine, the sole donor of O-GlcNAcylation produced by the hexosamine biosynthesis pathway, is controlled by the input of glucose, glutamine, acetyl coenzyme A and uridine triphosphate, making it a sensor of the fluctuation of molecules, making O-GlcNAcylation a pivotal nutrient sensor for the metabolism of carbohydrates, amino acids, lipids and nucleotides. O-GlcNAcylation, particularly prevalent in liver, is the core hub for controlling systemic glucose homeostasis due to its nutritional sensitivity and precise spatiotemporal regulation of insulin signal transduction. The pathology of various liver diseases has highlighted hepatic metabolic disorder and dysfunction, and abnormal O-GlcNAcylation also plays a specific pathological role in these processes. Therefore, this review describes the unique features of O-GlcNAcylation and its dynamic homeostasis maintenance. Additionally, it explains the underlying nutritional sensitivity of O-GlcNAcylation and discusses its mechanism of spatiotemporal modulation of insulin signal transduction and liver metabolic homeostasis during the fasting and feeding cycle. This review emphasises the pathophysiological implications of O-GlcNAcylation in nonalcoholic fatty liver disease, nonalcoholic steatohepatitis and hepatic fibrosis, and focuses on the adverse effects of hyper O-GlcNAcylation on liver cancer progression and metabolic reprogramming.
Collapse
Affiliation(s)
- Ya-Jie Hu
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xu Zhang
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hong-Ming Lv
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yang Liu
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shi-Ze Li
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
18
|
Kim MH, Lee JH, Lee JS, Kim DC, Yang JW, An HJ, Na JM, Jung WJ, Song DH. Perilipin1 Expression as a Prognostic Factor in Patients with Squamous Cell Carcinoma of the Lung. Diagnostics (Basel) 2023; 13:3475. [PMID: 37998612 PMCID: PMC10670494 DOI: 10.3390/diagnostics13223475] [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: 11/03/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
Perilipin (PLIN) is a major structural protein located on the surface of lipid droplets. PLIN plays an important role in human metabolism and is associated with metabolic diseases, such as obesity, diabetes, hypertension, and endocrine disorders. The dysregulation of lipid metabolism is one of the most prominent metabolic changes observed in cancers. Therefore, the PLIN protein family has recently attracted attention owing to its role in lipid metabolism and cancer. To date, no studies have addressed the association between the prognosis of lung cancer and PLIN1 expression. For the first time, we found that high PLIN1 expression was significantly correlated with worse disease-free survival (DFS) in lung squamous cell carcinoma (SCC). We examined PLIN1 expression by the immunohistochemical analysis of surgical lung SCC specimens obtained from 94 patients. We analyzed the correlation between PLIN1 expression, clinicopathological data, and patient survival, using a chi-squared test, Kaplan-Meier analysis with log-rank tests, and the multivariate Cox proportional hazards regression test. High PLIN1 expression was significantly correlated with lower DFS in the Kaplan-Meier analysis and the multivariate Cox proportional hazards regression model. High PLIN1 expression was significantly correlated with worse prognosis in lung SCC.
Collapse
Affiliation(s)
- Min Hye Kim
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
| | - Jeong Hee Lee
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jong Sil Lee
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Dong Chul Kim
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jung Wook Yang
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Hyo Jung An
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Pathology, Gyeongsang National University Changwon Hospital, Changwon 51472, Republic of Korea
| | - Ji Min Na
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Wook Jae Jung
- Department of Pathology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (M.H.K.)
| | - Dae Hyun Song
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Pathology, Gyeongsang National University Changwon Hospital, Changwon 51472, Republic of Korea
| |
Collapse
|
19
|
Muñoz JP, Basei FL, Rojas ML, Galvis D, Zorzano A. Mechanisms of Modulation of Mitochondrial Architecture. Biomolecules 2023; 13:1225. [PMID: 37627290 PMCID: PMC10452872 DOI: 10.3390/biom13081225] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrial network architecture plays a critical role in cellular physiology. Indeed, alterations in the shape of mitochondria upon exposure to cellular stress can cause the dysfunction of these organelles. In this scenario, mitochondrial dynamics proteins and the phospholipid composition of the mitochondrial membrane are key for fine-tuning the modulation of mitochondrial architecture. In addition, several factors including post-translational modifications such as the phosphorylation, acetylation, SUMOylation, and o-GlcNAcylation of mitochondrial dynamics proteins contribute to shaping the plasticity of this architecture. In this regard, several studies have evidenced that, upon metabolic stress, mitochondrial dynamics proteins are post-translationally modified, leading to the alteration of mitochondrial architecture. Interestingly, several proteins that sustain the mitochondrial lipid composition also modulate mitochondrial morphology and organelle communication. In this context, pharmacological studies have revealed that the modulation of mitochondrial shape and function emerges as a potential therapeutic strategy for metabolic diseases. Here, we review the factors that modulate mitochondrial architecture.
Collapse
Affiliation(s)
- Juan Pablo Muñoz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Fernanda Luisa Basei
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas, 13083-871 Campinas, SP, Brazil
| | - María Laura Rojas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - David Galvis
- Programa de Química Farmacéutica, Universidad CES, Medellín 050031, Colombia
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
20
|
Shin H, Leung A, Costello KR, Senapati P, Kato H, Moore RE, Lee M, Lin D, Tang X, Pirrotte P, Bouman Chen Z, Schones DE. Inhibition of DNMT1 methyltransferase activity via glucose-regulated O-GlcNAcylation alters the epigenome. eLife 2023; 12:e85595. [PMID: 37470704 PMCID: PMC10390045 DOI: 10.7554/elife.85595] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/19/2023] [Indexed: 07/21/2023] Open
Abstract
The DNA methyltransferase activity of DNMT1 is vital for genomic maintenance of DNA methylation. We report here that DNMT1 function is regulated by O-GlcNAcylation, a protein modification that is sensitive to glucose levels, and that elevated O-GlcNAcylation of DNMT1 from high glucose environment leads to alterations to the epigenome. Using mass spectrometry and complementary alanine mutation experiments, we identified S878 as the major residue that is O-GlcNAcylated on human DNMT1. Functional studies in human and mouse cells further revealed that O-GlcNAcylation of DNMT1-S878 results in an inhibition of methyltransferase activity, resulting in a general loss of DNA methylation that preferentially occurs at partially methylated domains (PMDs). This loss of methylation corresponds with an increase in DNA damage and apoptosis. These results establish O-GlcNAcylation of DNMT1 as a mechanism through which the epigenome is regulated by glucose metabolism and implicates a role for glycosylation of DNMT1 in metabolic diseases characterized by hyperglycemia.
Collapse
Affiliation(s)
- Heon Shin
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
| | - Amy Leung
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
| | - Kevin R Costello
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
- Irell and Manella Graduate School of Biological Sciences, City of HopeDuarteUnited States
| | - Parijat Senapati
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
| | - Hiroyuki Kato
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
| | - Roger E Moore
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center DuarteDuarteUnited States
| | - Michael Lee
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
- Irell and Manella Graduate School of Biological Sciences, City of HopeDuarteUnited States
| | - Dimitri Lin
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
| | - Xiaofang Tang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
| | - Patrick Pirrotte
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center DuarteDuarteUnited States
- Cancer & Cell Biology Division, Translational Genomics Research InstitutePhoenixUnited States
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
- Irell and Manella Graduate School of Biological Sciences, City of HopeDuarteUnited States
| | - Dustin E Schones
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of HopeDuarteUnited States
- Irell and Manella Graduate School of Biological Sciences, City of HopeDuarteUnited States
| |
Collapse
|
21
|
Nakamoto A, Ohashi N, Sugawara L, Morino K, Ida S, Perry RJ, Sakuma I, Yanagimachi T, Fujita Y, Ugi S, Kume S, Shulman GI, Maegawa H. O-linked N-acetylglucosamine modification is essential for physiological adipose expansion induced by high-fat feeding. Am J Physiol Endocrinol Metab 2023; 325:E46-E61. [PMID: 37224467 PMCID: PMC10292976 DOI: 10.1152/ajpendo.00263.2022] [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: 10/05/2022] [Revised: 05/01/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
Adipose tissues accumulate excess energy as fat and heavily influence metabolic homeostasis. O-linked N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation), which involves the addition of N-acetylglucosamine to proteins by O-GlcNAc transferase (Ogt), modulates multiple cellular processes. However, little is known about the role of O-GlcNAcylation in adipose tissues during body weight gain due to overnutrition. Here, we report on O-GlcNAcylation in mice with high-fat diet (HFD)-induced obesity. Mice with knockout of Ogt in adipose tissue achieved using adiponectin promoter-driven Cre recombinase (Ogt-FKO) gained less body weight than control mice under HFD. Surprisingly, Ogt-FKO mice exhibited glucose intolerance and insulin resistance, despite their reduced body weight gain, as well as decreased expression of de novo lipogenesis genes and increased expression of inflammatory genes, resulting in fibrosis at 24 weeks of age. Primary cultured adipocytes derived from Ogt-FKO mice showed decreased lipid accumulation. Both primary cultured adipocytes and 3T3-L1 adipocytes treated with OGT inhibitor showed increased secretion of free fatty acids. Medium derived from these adipocytes stimulated inflammatory genes in RAW 264.7 macrophages, suggesting that cell-to-cell communication via free fatty acids might be a cause of adipose inflammation in Ogt-FKO mice. In conclusion, O-GlcNAcylation is important for healthy adipose expansion in mice. Glucose flux into adipose tissues may be a signal to store excess energy as fat.NEW & NOTEWORTHY We evaluated the role of O-GlcNAcylation in adipose tissue in diet-induced obesity using adipose tissue-specific Ogt knockout mice. We found that O-GlcNAcylation in adipose tissue is essential for healthy fat expansion and that Ogt-FKO mice exhibit severe fibrosis upon long-term overnutrition. O-GlcNAcylation in adipose tissue may regulate de novo lipogenesis and free fatty acid efflux to the degree of overnutrition. We believe that these results provide new insights into adipose tissue physiology and obesity research.
Collapse
Affiliation(s)
- Akiko Nakamoto
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Natsuko Ohashi
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Lucia Sugawara
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Katsutaro Morino
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
- Institutional Research Office, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shogo Ida
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Rachel J Perry
- Department of Medicine (Endocrinology), Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Ikki Sakuma
- Department of Medicine (Endocrinology), Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | | | - Yukihiro Fujita
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Satoshi Ugi
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Gerald I Shulman
- Department of Medicine (Endocrinology), Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
22
|
Biernacka KM, Giri D, Hawton K, Segers F, Perks CM, Hamilton-Shield JP. Case report: Molecular characterisation of adipose-tissue derived cells from a patient with ROHHAD syndrome. Front Pediatr 2023; 11:1128216. [PMID: 37456561 PMCID: PMC10348915 DOI: 10.3389/fped.2023.1128216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
There have been over 100 cases of Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation (ROHHAD) syndrome reported, but there is currently no curative treatment for children with this condition. We aimed to better characterise adipose cells from a child with ROHHAD syndrome. We isolated pre-adipocytes from a 4 year-old female patient with ROHHAD syndrome and assessed proliferation rate of these cells. We evaluated levels of DLP-Pref-1(pre-adipocyte marker) using western blotting, and concentrations of interleukin-6(IL-6) using ELISA. We performed next-generation sequencing (NGS) and bioinformatic analyses on these cells compared to tissue from an age/sex-matched control. The two most up-/down-regulated genes were validated using QPCR. We successfully isolated pre-adipocytes from a fat biopsy, by confirming the presence of Pref-1 and differentiated them to mature adipocytes. Interleukin 6, (Il-6) levels were 5.6-fold higher in ROHHAD cells compared to a control age/sex-matched biopsy. NGS revealed 25,703 differentially expressed genes (DEGs) from ROHHAD cells vs. control of which 2,237 genes were significantly altered. The 20 most significantly up/down-regulated genes were selected for discussion. This paper describes the first transcriptomic analysis of adipose cells from a child with ROHHAD vs. normal control adipose tissue as a first step in identifying targetable pathways/mechanisms underlying this condition with novel therapeutic interventions.
Collapse
Affiliation(s)
- Kalina M. Biernacka
- Cancer Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, Bristol, United Kingdom
| | - Dinesh Giri
- Department of Paediatric Endocrinology and Diabetes, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Katherine Hawton
- Department of Paediatric Endocrinology and Diabetes, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Francisca Segers
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - Claire M. Perks
- Cancer Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, Bristol, United Kingdom
| | - Julian P. Hamilton-Shield
- Department of Translational Health Sciences, Nutrition Theme, NIHR Bristol Biomedical Research Centre, Bristol Medical School, University of Bristol, UBHT Education Centre, Bristol, United Kingdom
| |
Collapse
|
23
|
Griffin ME, Thompson JW, Xiao Y, Sweredoski MJ, Aksenfeld RB, Jensen EH, Koldobskaya Y, Schacht AL, Kim TD, Choudhry P, Lomenick B, Garbis SD, Moradian A, Hsieh-Wilson LC. Functional glycoproteomics by integrated network assembly and partitioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.541482. [PMID: 37398272 PMCID: PMC10312638 DOI: 10.1101/2023.06.13.541482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The post-translational modification (PTM) of proteins by O-linked β-N-acetyl-D-glucosamine (O-GlcNAcylation) is widespread across the proteome during the lifespan of all multicellular organisms. However, nearly all functional studies have focused on individual protein modifications, overlooking the multitude of simultaneous O-GlcNAcylation events that work together to coordinate cellular activities. Here, we describe Networking of Interactors and SubstratEs (NISE), a novel, systems-level approach to rapidly and comprehensively monitor O-GlcNAcylation across the proteome. Our method integrates affinity purification-mass spectrometry (AP-MS) and site-specific chemoproteomic technologies with network generation and unsupervised partitioning to connect potential upstream regulators with downstream targets of O-GlcNAcylation. The resulting network provides a data-rich framework that reveals both conserved activities of O-GlcNAcylation such as epigenetic regulation as well as tissue-specific functions like synaptic morphology. Beyond O-GlcNAc, this holistic and unbiased systems-level approach provides a broadly applicable framework to study PTMs and discover their diverse roles in specific cell types and biological states.
Collapse
Affiliation(s)
- Matthew E. Griffin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Co-first author
| | - John W. Thompson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Co-first author
| | - Yao Xiao
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Co-first author
| | - Michael J. Sweredoski
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Rita B. Aksenfeld
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Elizabeth H. Jensen
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yelena Koldobskaya
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Andrew L. Schacht
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Terry D. Kim
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Priya Choudhry
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Brett Lomenick
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Spiros D. Garbis
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Annie Moradian
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Linda C. Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Lead contact
| |
Collapse
|
24
|
Wu X, Xu M, Geng M, Chen S, Little PJ, Xu S, Weng J. Targeting protein modifications in metabolic diseases: molecular mechanisms and targeted therapies. Signal Transduct Target Ther 2023; 8:220. [PMID: 37244925 PMCID: PMC10224996 DOI: 10.1038/s41392-023-01439-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/01/2023] [Accepted: 04/06/2023] [Indexed: 05/29/2023] Open
Abstract
The ever-increasing prevalence of noncommunicable diseases (NCDs) represents a major public health burden worldwide. The most common form of NCD is metabolic diseases, which affect people of all ages and usually manifest their pathobiology through life-threatening cardiovascular complications. A comprehensive understanding of the pathobiology of metabolic diseases will generate novel targets for improved therapies across the common metabolic spectrum. Protein posttranslational modification (PTM) is an important term that refers to biochemical modification of specific amino acid residues in target proteins, which immensely increases the functional diversity of the proteome. The range of PTMs includes phosphorylation, acetylation, methylation, ubiquitination, SUMOylation, neddylation, glycosylation, palmitoylation, myristoylation, prenylation, cholesterylation, glutathionylation, S-nitrosylation, sulfhydration, citrullination, ADP ribosylation, and several novel PTMs. Here, we offer a comprehensive review of PTMs and their roles in common metabolic diseases and pathological consequences, including diabetes, obesity, fatty liver diseases, hyperlipidemia, and atherosclerosis. Building upon this framework, we afford a through description of proteins and pathways involved in metabolic diseases by focusing on PTM-based protein modifications, showcase the pharmaceutical intervention of PTMs in preclinical studies and clinical trials, and offer future perspectives. Fundamental research defining the mechanisms whereby PTMs of proteins regulate metabolic diseases will open new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiumei Wu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, China
| | - Mengyun Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Mengya Geng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shuo Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya, QLD, 4575, Australia
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, China.
- Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
25
|
Wu HF, Huang CW, Art J, Liu HX, Hart GW, Zeltner N. O-GlcNAcylation is crucial for sympathetic neuron development, maintenance, functionality and contributes to peripheral neuropathy. Front Neurosci 2023; 17:1137847. [PMID: 37229433 PMCID: PMC10203903 DOI: 10.3389/fnins.2023.1137847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/12/2023] [Indexed: 05/27/2023] Open
Abstract
O-GlcNAcylation is a post-translational modification (PTM) that regulates a wide range of cellular functions and has been associated with multiple metabolic diseases in various organs. The sympathetic nervous system (SNS) is the efferent portion of the autonomic nervous system that regulates metabolism of almost all organs in the body. How much the development and functionality of the SNS are influenced by O-GlcNAcylation, as well as how such regulation could contribute to sympathetic neuron (symN)-related neuropathy in diseased states, remains unknown. Here, we assessed the level of protein O-GlcNAcylation at various stages of symN development, using a human pluripotent stem cell (hPSC)-based symN differentiation paradigm. We found that pharmacological disruption of O-GlcNAcylation impaired both the growth and survival of hPSC-derived symNs. In the high glucose condition that mimics hyperglycemia, hPSC-derived symNs were hyperactive, and their regenerative capacity was impaired, which resembled typical neuronal defects in patients and animal models of diabetes mellitus. Using this model of sympathetic neuropathy, we discovered that O-GlcNAcylation increased in symNs under high glucose, which lead to hyperactivity. Pharmacological inhibition of O-GlcNAcylation rescued high glucose-induced symN hyperactivity and cell stress. This framework provides the first insight into the roles of O-GlcNAcylation in both healthy and diseased human symNs and may be used as a platform for therapeutic studies.
Collapse
Affiliation(s)
- Hsueh-Fu Wu
- Center for Molecular Medicine, University of Georgia, Athens, GA, United States
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
| | - Chia-Wei Huang
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Jennifer Art
- Center for Molecular Medicine, University of Georgia, Athens, GA, United States
- Biomedical and Translational Sciences Institute, Neuroscience Program, University of Georgia, Athens, GA, United States
| | - Hong-Xiang Liu
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Gerald W. Hart
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, GA, United States
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| |
Collapse
|
26
|
Zhang X, Ha S, Lau HCH, Yu J. Excess body weight: Novel insights into its roles in obesity comorbidities. Semin Cancer Biol 2023; 92:16-27. [PMID: 36965839 DOI: 10.1016/j.semcancer.2023.03.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Excess body weight is a global health problem due to sedentary lifestyle and unhealthy diet, affecting 2 billion population worldwide. Obesity is a major risk factor for metabolic diseases. Notably, the metabolic risk of obesity largely depends on body weight distribution, of which visceral adipose tissues but not subcutaneous fats are closely associated with obesity comorbidities, including type 2 diabetes, non-alcoholic fatty liver disease, cardiovascular disease and certain types of cancer. Latest multi-omics and mechanistical studies reported the crucial involvement of genetic and epigenetic alterations, adipokines dysregulation, immunity changes, imbalance of white and brown adipose tissues, and gut microbial dysbiosis in mediating the pathogenic association between visceral adipose tissues and comorbidities. In this review, we explore the epidemiology of excess body weight and the up-to-date mechanism of how excess body weight and obesity lead to chronic complications. We also examine the utilization of visceral fat measurement as an accurate clinical parameter for risk assessment in healthy individuals and clinical outcome prediction in obese subjects. In addition, current approaches for the prevention and treatment of excess body weight and its related metabolic comorbidities are further discussed. DATA AVAILABILITY: No data was used for the research described in the article.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Suki Ha
- Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
27
|
Adila F, Rejeki PS, Herawati L. Acute Moderate-Intensity Strength Exercise Increases Anti-Inflammatory Cytokines in Obese Females. PHYSICAL EDUCATION THEORY AND METHODOLOGY 2023; 23:35-41. [DOI: 10.17309/tmfv.2023.1.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The study purpose was to prove the effect of acute moderate-intensity endurance and strength exercise on increasing IL-6 levels in obese females.
Materials and methods. A total of 21 obese women aged 20-25 years were recruited from among university students and given two modes of acute exercise intervention, namely moderate-intensity endurance and strength exercise carried out for 35 minutes/session. Subjects were divided randomly into three groups, namely K1 (control group without intervention; n = 7), K2 (Acute moderate-intensity endurance exercise; n = 7), K3 (Acute moderate-intensity strength exercise; n = 7). ELISA was used to analyze serum IL-6 levels before and after exercise. The data analysis technique used the One-way ANOVA test and continued with the Tukey HSD post-hoc test with a significance level of 5%.
Results. The results of the One-way ANOVA test showed that there was a significant difference between serum IL-6 levels after exercise and delta (Δ) in the three groups (p ≤ 0.01). The results of the Tukey HSD post-hoc test showed that there was a significant difference between serum IL-6 levels after exercise and delta (Δ) at K3 with K1 (p ≤ 0.01), K3with K2 (p ≤ 0.01), while there was no significant difference in serum IL-6 levels (p ≥ 0.05) at K2 with K1.
Conclusions. Overall, our study concluded that 35 min/session of acute moderate-intensity strength exercise was effective in increasing anti-inflammatory cytokines, such as IL-6, in obese females.
Collapse
|
28
|
Chen X, Yong H, Chen M, Deng C, Wang P, Chu S, Li M, Hou P, Zheng J, Li Z, Bai J. TRIM21 attenuates renal carcinoma lipogenesis and malignancy by regulating SREBF1 protein stability. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:34. [PMID: 36694250 PMCID: PMC9875457 DOI: 10.1186/s13046-022-02583-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/24/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Metabolic reprogramming is a hallmark of various cancers. Targeting metabolic processes is a very attractive treatment for cancer. Renal cell carcinoma (RCC) is a type of metabolic disease, and the lipidomic profile of RCC is significantly altered compared with that of healthy tissue. However, the molecular mechanism underlying lipid metabolism regulation in RCC is not clear. METHODS The XF long-chain fatty acid oxidative stress test kits were used to assess the dependence on long-chain fatty acids and mitochondrial function after knockdown TRIM21 in RCC cells. The effect of TRIM21 on the lipid content in RCC cells was determined by metabolomics analysis, Oil Red O staining, and cellular Nile red staining. qRT-PCR and western blot were used to explore the relationship between TRIM21 and lipogenesis, and then the key molecule sterol regulatory element binding transcription factor 1 (SREBF1) was identified to interact with TRIM21 by immunoprecipitation, which was also identified in an orthotopic model. Subsequently, the relevance and clinical significance of TRIM21 and SREBF1 were analyzed by The Cancer Genome Atlas (TCGA) database, and 239 tissues were collected from RCC patients. RESULTS TRIM21 silencing attenuated the dependence of RCC cells on fatty acids, and enhanced lipid accumulation in RCC cells. TRIM21 overexpression significantly decreased lipid contents by decreasing the expression of lipogenic enzymes via ubiquitination-mediated degradation of SREBF1. SREBF1 is critical for TRIM21-mediated lipogenesis inhibition in vitro and in vivo. Moreover, TRIM21 expression is negatively correlated with SREBF1 expression, and TRIM21-SREBF1 is a reliable combinational biomarker for RCC prognosis. CONCLUSION The findings from this study reveal a novel pathway through which TRIM21 inhibits the lipid metabolism process of RCC and shed light on the development of targeted metabolic treatment and prognosis diagnosis of RCC.
Collapse
Affiliation(s)
- Xintian Chen
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China ,grid.413389.40000 0004 1758 1622Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Xuzhou, China ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu 221004 Xuzhou, China
| | - Hongmei Yong
- grid.417303.20000 0000 9927 0537Department of Oncology, The Second People’s Hospital of Huai’an, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huaian, Jiangsu China
| | - Miaolei Chen
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China
| | - Chuyin Deng
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China
| | - Pengfei Wang
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China
| | - Sufang Chu
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China
| | - Minle Li
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China ,grid.413389.40000 0004 1758 1622Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Xuzhou, China ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu 221004 Xuzhou, China
| | - Pingfu Hou
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China ,grid.413389.40000 0004 1758 1622Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Xuzhou, China ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu 221004 Xuzhou, China
| | - Junnian Zheng
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China ,grid.413389.40000 0004 1758 1622Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Xuzhou, China ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu 221004 Xuzhou, China
| | - Zhongwei Li
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China ,grid.413389.40000 0004 1758 1622Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Xuzhou, China ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu 221004 Xuzhou, China
| | - Jin Bai
- grid.417303.20000 0000 9927 0537Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu Province 221004 Xuzhou, China ,grid.413389.40000 0004 1758 1622Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Xuzhou, China ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Jiangsu 221004 Xuzhou, China
| |
Collapse
|
29
|
Yu Y, Zhang YH, Liu L, Yu LL, Li JP, Rao JA, Hu F, Zhu LJ, Bao HH, Cheng XS. Bioinformatics analysis of candidate genes and potential therapeutic drugs targeting adipose tissue in obesity. Adipocyte 2022; 11:1-10. [PMID: 34964707 PMCID: PMC8726706 DOI: 10.1080/21623945.2021.2013406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Obesity is a complex medical condition that affects multiple organs in the body. However, the underlying mechanisms of obesity, as well as its treatment, are largely unexplored. The focus of this research was to use bioinformatics to discover possible treatment targets for obesity. To begin, the GSE133099 database was used to identify 364 differentially expressed genes (DEGs). Then, DEGs were subjected to tissue-specific analyses and enrichment analyses, followed by the creation of a protein-protein interaction (PPI) network and generation of a drug-gene interaction database to screen key genes and potential future drugs targeting obesity. Findings have illustrated that the tissue-specific expression of neurologic markers varied significantly (34.7%, 52/150). Among these genes, Lep, ApoE, Fyn, and FN1 were the key genes observed in the adipocyte samples from obese patients relative to the controls. Furthermore, nine potential therapeutic drugs (dasatinib, ocriplasmin, risperidone, gemfibrozil, ritonavir, fluvastatin, pravastatin, warfarin, atorvastatin) that target the key genes were also screened and selected. To conclude the key genes discovered (Lep, ApoE, Fyn, and FN1), as well as 9 candidate drugs, could be used as therapeutic targets in treating obesity.
Collapse
Affiliation(s)
- Yun Yu
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu-Han Zhang
- Reproductive Medical Center, Maternal and Child Health Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liang Liu
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ling-Ling Yu
- Department of Rehabilitation, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun-Pei Li
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing-an Rao
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Hu
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ling-Juan Zhu
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui-Hui Bao
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-Shu Cheng
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Hu A, Zou H, Chen B, Zhong J. Posttranslational modifications in diabetes: Mechanisms and functions. Rev Endocr Metab Disord 2022; 23:1011-1033. [PMID: 35697961 DOI: 10.1007/s11154-022-09740-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 12/15/2022]
Abstract
As one of the most widespread chronic diseases, diabetes and its accompanying complications affect approximately one tenth of individuals worldwide and represent a growing cause of morbidity and mortality. Accumulating evidence has proven that the process of diabetes is complex and interactive, involving various cellular responses and signaling cascades by posttranslational modifications (PTMs). Therefore, understanding the mechanisms and functions of PTMs in regulatory networks has fundamental importance for understanding the prediction, onset, diagnosis, progression, and treatment of diabetes. In this review, we offer a holistic summary and illustration of the crosstalk between PTMs and diabetes, including both types 1 and 2. Meanwhile, we discuss the potential use of PTMs in diabetes treatment and provide a prospective direction for deeply understanding the metabolic diseases.
Collapse
Affiliation(s)
- Ang Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China
| | - Haohong Zou
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China
| | - Bin Chen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jianing Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
31
|
Diversity of change in body mass index and skinfold thickness between different study courses within four years of study among the male students in a university in Poland. ANTHROPOLOGICAL REVIEW 2022. [DOI: 10.18778/1898-6773.85.3.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective: This study examined whether there were significant changes (expectedly increase) in BMI (Body Mass Index), and relative subcutaneous body fat during the four years of study in a university and compared these changes between the students of different courses, viz., Computer science, Law and administration, Humanities and Physical education.
Design: This was a prospective follow up study with measurements at two time points with gap of four years. Body mass index, Triceps, subscapular, mid-axillary, abdominal, supra-illiac and medial-calf skinfold thicknesses were measured at two points of time, just after entrance to university and again after completion of four years. Student’s T-test, one-way ANOVA, and repeated measures ANNOVA (two-way) were employed to assess significance of differences in anthropometric measures between groups of students.
Setting: The study was conducted at the University of Rzeszów, Poland.
Participants: 191 young men university students aged approximately 19.5 years and 24.0 years, at the beginning and after four years, respectively, during this study.
Results: Students of all courses underwent increments in BMI, absolute- and relative skinfold thicknesses, except that the students of physical education course did not show change in skinfold thicknesses relative to BMI.
Conclusions: It has been concluded that the change in the adiposity profile during the years of study at university varied according to the course types. Further, detailed studies on the nature and cause of such variation occurring between course types may lead to better understanding etiology of overweight and obesity before entering to adult life.
Collapse
|
32
|
The Novel Peptide Chm-273s Has Therapeutic Potential for Metabolic Disorders: Evidence from In Vitro Studies and High-Sucrose Diet and High-Fat Diet Rodent Models. Pharmaceutics 2022; 14:pharmaceutics14102088. [PMID: 36297523 PMCID: PMC9611607 DOI: 10.3390/pharmaceutics14102088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to develop a novel peptide potentially applicable for the treatment of metabolic conditions, such as obesity and type 2 diabetes (T2D). We identified CHM-273S from the list of peptides from milk hydrolysate obtained by HPLC/MS-MS. In vitro analysis of primary murine fibroblasts indicated the potential of CHM-273S to upregulate IRS2 mRNA expression. CHM-273S showed a prominent anorexigenic effect in mice with the induction of a key mechanism of leptin signaling via STAT3 in the hypothalamus as a possible effector. In the animal model of metabolic disease, CHM-273S alleviated glucose intolerance and insulin resistance, and induced phosphorylation of Akt at Ser473 and Thr308 in the hepatocytes of high-sucrose diet-fed rats. In a murine model of T2D, CHM-273S mitigated high-fat diet-induced hyperglycemia and insulin resistance and improved low-grade inflammation by diminishing serum TNFα. Mice treated with chronic CHM-273S had a significant reduction in body weight, with a lower visceral fat pad weight and narrow adipocytes. The effects of the peptide administration were comparable to those of metformin. We show the potential of CHM-273S to alleviate diet-induced metabolic alterations in rodents, substantiating its further development as a therapeutic for obesity, T2D, and other metabolic conditions.
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
|
Liu Y, Hu Y, Li S. Protein O-GlcNAcylation in Metabolic Modulation of Skeletal Muscle: A Bright but Long Way to Go. Metabolites 2022; 12:888. [PMID: 36295790 PMCID: PMC9610910 DOI: 10.3390/metabo12100888] [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: 08/19/2022] [Revised: 09/09/2022] [Accepted: 09/17/2022] [Indexed: 09/07/2024] Open
Abstract
O-GlcNAcylation is an atypical, dynamic and reversible O-glycosylation that is critical and abundant in metazoan. O-GlcNAcylation coordinates and receives various signaling inputs such as nutrients and stresses, thus spatiotemporally regulating the activity, stability, localization and interaction of target proteins to participate in cellular physiological functions. Our review discusses in depth the involvement of O-GlcNAcylation in the precise regulation of skeletal muscle metabolism, such as glucose homeostasis, insulin sensitivity, tricarboxylic acid cycle and mitochondrial biogenesis. The complex interaction and precise modulation of O-GlcNAcylation in these nutritional pathways of skeletal muscle also provide emerging mechanical information on how nutrients affect health, exercise and disease. Meanwhile, we explored the potential role of O-GlcNAcylation in skeletal muscle pathology and focused on its benefits in maintaining proteostasis under atrophy. In general, these understandings of O-GlcNAcylation are conducive to providing new insights into skeletal muscle (patho) physiology.
Collapse
Affiliation(s)
| | | | - Shize Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| |
Collapse
|
35
|
Protein O-GlcNAcylation and the regulation of energy homeostasis: lessons from knock-out mouse models. J Biomed Sci 2022; 29:64. [PMID: 36058931 PMCID: PMC9443036 DOI: 10.1186/s12929-022-00851-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
O-GlcNAcylation corresponds to the addition of N-Acetylglucosamine (GlcNAc) on serine or threonine residues of cytosolic, nuclear and mitochondrial proteins. This reversible modification is catalysed by a unique couple of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). OGT uses UDP-GlcNAc produced in the hexosamine biosynthesis pathway, to modify proteins. UDP-GlcNAc is at the cross-roads of several cellular metabolisms, including glucose, amino acids and fatty acids. Therefore, OGT is considered as a metabolic sensor that post-translationally modifies proteins according to nutrient availability. O-GlcNAcylation can modulate protein–protein interactions and regulate protein enzymatic activities, stability or subcellular localization. In addition, it can compete with phosphorylation on the same serine or threonine residues, or regulate positively or negatively the phosphorylation of adjacent residues. As such, O-GlcNAcylation is a major actor in the regulation of cell signaling and has been implicated in numerous physiological and pathological processes. A large body of evidence have indicated that increased O-GlcNAcylation participates in the deleterious effects of glucose (glucotoxicity) in metabolic diseases. However, recent studies using mice models with OGT or OGA knock-out in different tissues have shown that O-GlcNAcylation protects against various cellular stresses, and indicate that both increase and decrease in O-GlcNAcylation have deleterious effects on the regulation of energy homeostasis.
Collapse
|
36
|
Wang Q, Zhang B, Stutz B, Liu ZW, Horvath TL, Yang X. Ventromedial hypothalamic OGT drives adipose tissue lipolysis and curbs obesity. SCIENCE ADVANCES 2022; 8:eabn8092. [PMID: 36044565 PMCID: PMC9432828 DOI: 10.1126/sciadv.abn8092] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/14/2022] [Indexed: 05/31/2023]
Abstract
The ventromedial hypothalamus (VMH) is known to regulate body weight and counterregulatory response. However, how VMH neurons regulate lipid metabolism and energy balance remains unknown. O-linked β-d-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation), catalyzed by O-GlcNAc transferase (OGT), is considered a cellular sensor of nutrients and hormones. Here, we report that genetic ablation of OGT in VMH neurons inhibits neuronal excitability. Mice with VMH neuron-specific OGT deletion show rapid weight gain, increased adiposity, and reduced energy expenditure, without significant changes in food intake or physical activity. The obesity phenotype is associated with adipocyte hypertrophy and reduced lipolysis of white adipose tissues. In addition, OGT deletion in VMH neurons down-regulates the sympathetic activity and impairs the sympathetic innervation of white adipose tissues. These findings identify OGT in the VMH as a homeostatic set point that controls body weight and underscore the importance of the VMH in regulating lipid metabolism through white adipose tissue-specific innervation.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Bichen Zhang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Bernardo Stutz
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L. Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiaoyong Yang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
37
|
Lockridge A, Hanover JA. A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne) 2022; 13:943576. [PMID: 36111295 PMCID: PMC9468787 DOI: 10.3389/fendo.2022.943576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although traditionally considered a glucose metabolism-associated modification, the O-linked β-N-Acetylglucosamine (O-GlcNAc) regulatory system interacts extensively with lipids and is required to maintain lipid homeostasis. The enzymes of O-GlcNAc cycling have molecular properties consistent with those expected of broad-spectrum environmental sensors. By direct protein-protein interactions and catalytic modification, O-GlcNAc cycling enzymes may provide both acute and long-term adaptation to stress and other environmental stimuli such as nutrient availability. Depending on the cell type, hyperlipidemia potentiates or depresses O-GlcNAc levels, sometimes biphasically, through a diversity of unique mechanisms that target UDP-GlcNAc synthesis and the availability, activity and substrate selectivity of the glycosylation enzymes, O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA). At the same time, OGT activity in multiple tissues has been implicated in the homeostatic regulation of systemic lipid uptake, storage and release. Hyperlipidemic patterns of O-GlcNAcylation in these cells are consistent with both transient physiological adaptation and feedback uninhibited obesogenic and metabolic dysregulation. In this review, we summarize the numerous interconnections between lipid and O-GlcNAc metabolism. These links provide insights into how the O-GlcNAc regulatory system may contribute to lipid-associated diseases including obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Amber Lockridge
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
38
|
Xu B, Zhang C, Jiang A, Zhang X, Liang F, Wang X, Li D, Liu C, Liu X, Xia J, Li Y, Wang Y, Yang Z, Chen J, Zhou Y, Chen L, Sun H. Histone methyltransferase Dot1L recruits O-GlcNAc transferase to target chromatin sites to regulate histone O-GlcNAcylation. J Biol Chem 2022; 298:102115. [PMID: 35690146 PMCID: PMC9283943 DOI: 10.1016/j.jbc.2022.102115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/05/2022] Open
Abstract
O-GlcNAc transferase (OGT) is the distinctive enzyme responsible for catalyzing O-GlcNAc addition to the serine or threonine residues of thousands of cytoplasmic and nuclear proteins involved in such basic cellular processes as DNA damage repair, RNA splicing, and transcription preinitiation and initiation complex assembly. However, the molecular mechanism by which OGT regulates gene transcription remains elusive. Using proximity labeling-based mass spectrometry, here, we searched for functional partners of OGT and identified interacting protein Dot1L, a conserved and unique histone methyltransferase known to mediate histone H3 Lys79 methylation, which is required for gene transcription, DNA damage repair, cell proliferation, and embryo development. Although this specific interaction with OGT does not regulate the enzymatic activity of Dot1L, we show that it does facilitate OGT-dependent histone O-GlcNAcylation. Moreover, we demonstrate that OGT associates with Dot1L at transcription start sites and that depleting Dot1L decreases OGT associated with chromatin globally. Notably, we also show that downregulation of Dot1L reduces the levels of histone H2B S112 O-GlcNAcylation and histone H2B K120 ubiquitination in vivo, which are associated with gene transcription regulation. Taken together, these results reveal that O-GlcNAcylation of chromatin is dependent on Dot1L.
Collapse
Affiliation(s)
- Bo Xu
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Can Zhang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Ao Jiang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Xianhong Zhang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Fenfei Liang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Xueqing Wang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Danni Li
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Chenglong Liu
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Xiaomei Liu
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Jing Xia
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Yang Li
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Yirong Wang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Zelan Yang
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Jia Chen
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Yu Zhou
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China
| | - Liang Chen
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China.
| | - Hui Sun
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei Province, China; Hubei Province key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430072, Hubei Province, China.
| |
Collapse
|
39
|
Liu Y, Hu YJ, Fan WX, Quan X, Xu B, Li SZ. O-GlcNAcylation: The Underestimated Emerging Regulators of Skeletal Muscle Physiology. Cells 2022; 11:1789. [PMID: 35681484 PMCID: PMC9180116 DOI: 10.3390/cells11111789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
O-GlcNAcylation is a highly dynamic, reversible and atypical glycosylation that regulates the activity, biological function, stability, sublocation and interaction of target proteins. O-GlcNAcylation receives and coordinates different signal inputs as an intracellular integrator similar to the nutrient sensor and stress receptor, which target multiple substrates with spatio-temporal analysis specifically to maintain cellular homeostasis and normal physiological functions. Our review gives a brief description of O-GlcNAcylation and its only two processing enzymes and HBP flux, which will help to better understand its physiological characteristics of sensing nutrition and environmental cues. This nutritional and stress-sensitive properties of O-GlcNAcylation allow it to participate in the precise regulation of skeletal muscle metabolism. This review discusses the mechanism of O-GlcNAcylation to alleviate metabolic disorders and the controversy about the insulin resistance of skeletal muscle. The level of global O-GlcNAcylation is precisely controlled and maintained in the "optimal zone", and its abnormal changes is a potential factor in the pathogenesis of cancer, neurodegeneration, diabetes and diabetic complications. Although the essential role of O-GlcNAcylation in skeletal muscle physiology has been widely studied and recognized, it still is underestimated and overlooked. This review highlights the latest progress and potential mechanisms of O-GlcNAcylation in the regulation of skeletal muscle contraction and structural properties.
Collapse
Affiliation(s)
| | | | | | | | - Bin Xu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (Y.L.); (Y.-J.H.); (W.-X.F.); (X.Q.)
| | - Shi-Ze Li
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (Y.L.); (Y.-J.H.); (W.-X.F.); (X.Q.)
| |
Collapse
|
40
|
Zhao M, Ren K, Xiong X, Xin Y, Zou Y, Maynard JC, Kim A, Battist AP, Koneripalli N, Wang Y, Chen Q, Xin R, Yang C, Huang R, Yu J, Huang Z, Zhang Z, Wang H, Wang D, Xiao Y, Salgado OC, Jarjour NN, Hogquist KA, Revelo XS, Burlingame AL, Gao X, von Moltke J, Lin Z, Ruan HB. Epithelial STAT6 O-GlcNAcylation drives a concerted anti-helminth alarmin response dependent on tuft cell hyperplasia and Gasdermin C. Immunity 2022; 55:623-638.e5. [PMID: 35385697 PMCID: PMC9109499 DOI: 10.1016/j.immuni.2022.03.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 12/14/2022]
Abstract
The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kaiqun Ren
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yue Xin
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yujie Zou
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jason C Maynard
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Angela Kim
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Alexander P Battist
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Navya Koneripalli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Yusu Wang
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Qianyue Chen
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Ruyue Xin
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chenyan Yang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Rong Huang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jiahui Yu
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zan Huang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Haiguang Wang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Daoyuan Wang
- College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Yihui Xiao
- College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Oscar C Salgado
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Nicholas N Jarjour
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xavier S Revelo
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Xiang Gao
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Zhaoyu Lin
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.
| |
Collapse
|
41
|
Role of Distinct Fat Depots in Metabolic Regulation and Pathological Implications. Rev Physiol Biochem Pharmacol 2022; 186:135-176. [PMID: 35915363 DOI: 10.1007/112_2022_73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
People suffering from obesity and associated metabolic disorders including diabetes are increasing exponentially around the world. Adipose tissue (AT) distribution and alteration in their biochemical properties play a major role in the pathogenesis of these diseases. Emerging evidence suggests that AT heterogeneity and depot-specific physiological changes are vital in the development of insulin resistance in peripheral tissues like muscle and liver. Classically, AT depots are classified into white adipose tissue (WAT) and brown adipose tissue (BAT); WAT is the site of fatty acid storage, while BAT is a dedicated organ of metabolic heat production. The discovery of beige adipocyte clusters in WAT depots indicates AT heterogeneity has a more central role than hither to ascribed. Therefore, we have discussed in detail the current state of understanding on cellular and molecular origin of different AT depots and their relevance toward physiological metabolic homeostasis. A major focus is to highlight the correlation between altered WAT distribution in the body and metabolic pathogenesis in animal models and humans. We have also underscored the disparity in the molecular (including signaling) changes in various WAT tissues during diabetic pathogenesis. Exercise-mediated beneficial alteration in WAT physiology/distribution that protects against metabolic disorders is evolving. Here we have discussed the depot-specific biochemical adjustments induced by different forms of exercise. A detailed understanding of the molecular details of inter-organ crosstalk via substrate utilization/storage and signaling through chemokines provide strategies to target selected WAT depots to pharmacologically mimic the benefits of exercise countering metabolic diseases including diabetes.
Collapse
|
42
|
Gonzalez-Rellan MJ, Fondevila MF, Dieguez C, Nogueiras R. O-GlcNAcylation: A Sweet Hub in the Regulation of Glucose Metabolism in Health and Disease. Front Endocrinol (Lausanne) 2022; 13:873513. [PMID: 35527999 PMCID: PMC9072661 DOI: 10.3389/fendo.2022.873513] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/23/2022] [Indexed: 12/17/2022] Open
Abstract
O-GlcNAcylation is a posttranslational modification ruled by the activity of a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). These two enzymes carry out the dynamic cycling of O-GlcNAcylation on a wide range of cytosolic, nuclear, and mitochondrial proteins in a nutrient- and stress-responsive manner. To maintain proper glucose homeostasis, a precise mechanism to sense blood glucose levels is required, to adapt cell physiology to fluctuations in nutrient intake to maintain glycemia within a narrow range. Disruptions in glucose homeostasis generates metabolic syndrome and type 2 diabetes. In this review we will discuss and summarize emerging findings that points O-GlcNAcylation as a hub in the control of systemic glucose homeostasis, and its involvement in the generation of insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Maria J. Gonzalez-Rellan
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- *Correspondence: Maria J. Gonzalez-Rellan, ; Marcos F. Fondevila,
| | - Marcos F. Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Maria J. Gonzalez-Rellan, ; Marcos F. Fondevila,
| | - Carlos Dieguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
43
|
Tools, tactics and objectives to interrogate cellular roles of O-GlcNAc in disease. Nat Chem Biol 2022; 18:8-17. [PMID: 34934185 PMCID: PMC8712397 DOI: 10.1038/s41589-021-00903-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/21/2021] [Indexed: 12/15/2022]
Abstract
The vast array of cell types of multicellular organisms must individually fine-tune their internal metabolism. One important metabolic and stress regulatory mechanism is the dynamic attachment/removal of glucose-derived sugar N-acetylglucosamine on proteins (O-GlcNAcylation). The number of proteins modified by O-GlcNAc is bewildering, with at least 7,000 sites in human cells. The outstanding challenge is determining how key O-GlcNAc sites regulate a target pathway amidst thousands of potential global sites. Innovative solutions are required to address this challenge in cell models and disease therapy. This Perspective shares critical suggestions for the O-GlcNAc field gleaned from the international O-GlcNAc community. Further, we summarize critical tools and tactics to enable newcomers to O-GlcNAc biology to drive innovation at the interface of metabolism and disease. The growing pace of O-GlcNAc research makes this a timely juncture to involve a wide array of scientists and new toolmakers to selectively approach the regulatory roles of O-GlcNAc in disease.
Collapse
|
44
|
Bai J, Li J, Liu N, Jia H, Si X, Zhai Z, Zhou Y, Yang Y, Ren F, Wu Z. Glucosamine alleviates zearalenone damage to porcine trophectoderm cells by activating PI3K/AKT signaling pathway. Food Funct 2022; 13:7857-7870. [DOI: 10.1039/d2fo00928e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
As one of the mycotoxins commonly found in feed and food, zearalenone (ZEA) mainly harms the reproductive functions of humans and animals. In our study, we investigated the protective effects...
Collapse
|
45
|
Dai W, Choubey M, Patel S, Singer HA, Ozcan L. Adipocyte CAMK2 deficiency improves obesity-associated glucose intolerance. Mol Metab 2021; 53:101300. [PMID: 34303021 PMCID: PMC8365526 DOI: 10.1016/j.molmet.2021.101300] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Obesity-related adipose tissue dysfunction has been linked to the development of insulin resistance, type 2 diabetes, and cardiovascular disease. Impaired calcium homeostasis is associated with altered adipose tissue metabolism; however, the molecular mechanisms that link disrupted calcium signaling to metabolic regulation are largely unknown. Here, we investigated the contribution of a calcium-sensing enzyme, calcium/calmodulin-dependent protein kinase II (CAMK2), to adipocyte function, obesity-associated insulin resistance, and glucose intolerance. METHODS To determine the impact of adipocyte CAMK2 deficiency on metabolic regulation, we generated a conditional knockout mouse model and acutely deleted CAMK2 in mature adipocytes. We further used in vitro differentiated adipocytes to dissect the mechanisms by which CAMK2 regulates adipocyte function. RESULTS CAMK2 activity was increased in obese adipose tissue, and depletion of adipocyte CAMK2 in adult mice improved glucose intolerance and insulin resistance without an effect on body weight. Mechanistically, we found that activation of CAMK2 disrupted adipocyte insulin signaling and lowered the amount of insulin receptor. Further, our results revealed that CAMK2 contributed to adipocyte lipolysis, tumor necrosis factor alpha (TNFα)-induced inflammation, and insulin resistance. CONCLUSIONS These results identify a new link between adipocyte CAMK2 activity, metabolic regulation, and whole-body glucose homeostasis.
Collapse
Affiliation(s)
- Wen Dai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mayank Choubey
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Sonal Patel
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
46
|
Pancreatic β-Cell O-GlcNAc Transferase Overexpression Increases Susceptibility to Metabolic Stressors in Female Mice. Cells 2021; 10:cells10102801. [PMID: 34685781 PMCID: PMC8534823 DOI: 10.3390/cells10102801] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 01/21/2023] Open
Abstract
The nutrient-sensor O-GlcNAc transferase (Ogt), the sole enzyme that adds an O-GlcNAc-modification onto proteins, plays a critical role for pancreatic β-cell survival and insulin secretion. We hypothesized that β-cell Ogt overexpression would confer protection from β-cell failure in response to metabolic stressors, such as high-fat diet (HFD) and streptozocin (STZ). Here, we generated a β-cell-specific Ogt in overexpressing (βOgtOE) mice, where a significant increase in Ogt protein level and O-GlcNAc-modification of proteins were observed in islets under a normal chow diet. We uncovered that βOgtOE mice show normal peripheral insulin sensitivity and glucose tolerance with a regular chow diet. However, when challenged with an HFD, only female βOgtOE (homozygous) Hz mice developed a mild glucose intolerance, despite increased insulin secretion and normal β-cell mass. While female mice are normally resistant to low-dose STZ treatments, the βOgtOE Hz mice developed hyperglycemia and glucose intolerance post-STZ treatment. Transcriptome analysis between islets with loss or gain of Ogt by RNA sequencing shows common altered pathways involving pro-survival Erk and Akt and inflammatory regulators IL1β and NFkβ. Together, these data show a possible gene dosage effect of Ogt and the importance O-GlcNAc cycling in β-cell survival and function to regulate glucose homeostasis.
Collapse
|
47
|
Nishimura Y, Yamakawa D, Uchida K, Shiromizu T, Watanabe M, Inagaki M. Primary cilia and lipid raft dynamics. Open Biol 2021; 11:210130. [PMID: 34428960 PMCID: PMC8385361 DOI: 10.1098/rsob.210130] [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] [Indexed: 01/13/2023] Open
Abstract
Primary cilia, antenna-like structures of the plasma membrane, detect various extracellular cues and transduce signals into the cell to regulate a wide range of functions. Lipid rafts, plasma membrane microdomains enriched in cholesterol, sphingolipids and specific proteins, are also signalling hubs involved in a myriad of physiological functions. Although impairment of primary cilia and lipid rafts is associated with various diseases, the relationship between primary cilia and lipid rafts is poorly understood. Here, we review a newly discovered interaction between primary cilia and lipid raft dynamics that occurs during Akt signalling in adipogenesis. We also discuss the relationship between primary cilia and lipid raft-mediated Akt signalling in cancer biology. This review provides a novel perspective on primary cilia in the regulation of lipid raft dynamics.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Daishi Yamakawa
- Department of Physiology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Katsunori Uchida
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takashi Shiromizu
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Masatoshi Watanabe
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Masaki Inagaki
- Department of Physiology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
48
|
Mammalian cell proliferation requires noncatalytic functions of O-GlcNAc transferase. Proc Natl Acad Sci U S A 2021; 118:2016778118. [PMID: 33419956 DOI: 10.1073/pnas.2016778118] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
O-GlcNAc transferase (OGT), found in the nucleus and cytoplasm of all mammalian cell types, is essential for cell proliferation. Why OGT is required for cell growth is not known. OGT performs two enzymatic reactions in the same active site. In one, it glycosylates thousands of different proteins, and in the other, it proteolytically cleaves another essential protein involved in gene expression. Deconvoluting OGT's myriad cellular roles has been challenging because genetic deletion is lethal; complementation methods have not been established. Here, we developed approaches to replace endogenous OGT with separation-of-function variants to investigate the importance of OGT's enzymatic activities for cell viability. Using genetic complementation, we found that OGT's glycosyltransferase function is required for cell growth but its protease function is dispensable. We next used complementation to construct a cell line with degron-tagged wild-type OGT. When OGT was degraded to very low levels, cells stopped proliferating but remained viable. Adding back catalytically inactive OGT rescued growth. Therefore, OGT has an essential noncatalytic role that is necessary for cell proliferation. By developing a method to quantify how OGT's catalytic and noncatalytic activities affect protein abundance, we found that OGT's noncatalytic functions often affect different proteins from its catalytic functions. Proteins involved in oxidative phosphorylation and the actin cytoskeleton were especially impacted by the noncatalytic functions. We conclude that OGT integrates both catalytic and noncatalytic functions to control cell physiology.
Collapse
|
49
|
Lockridge A, Jo S, Gustafson E, Damberg N, Mohan R, Olson M, Abrahante JE, Alejandro EU. Islet O-GlcNAcylation Is Required for Lipid Potentiation of Insulin Secretion through SERCA2. Cell Rep 2021; 31:107609. [PMID: 32375037 DOI: 10.1016/j.celrep.2020.107609] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 01/24/2020] [Accepted: 04/12/2020] [Indexed: 12/19/2022] Open
Abstract
During early obesity, pancreatic β cells compensate for increased metabolic demand through a transient phase of insulin hypersecretion that stabilizes blood glucose and forestalls diabetic progression. We find evidence that β cell O-GlcNAcylation, a nutrient-responsive post-translational protein modification regulated by O-GlcNAc transferase (OGT), is critical for coupling hyperlipidemia to β cell functional adaptation during this compensatory prediabetic phase. In mice, islet O-GlcNAcylation rises and falls in tandem with the timeline of secretory potentiation during high-fat feeding while genetic models of β-cell-specific OGT loss abolish hyperinsulinemic responses to lipids, in vivo and in vitro. We identify the endoplasmic reticulum (ER) Ca2+ ATPase SERCA2 as a β cell O-GlcNAcylated protein in mice and humans that is able to rescue palmitate-stimulated insulin secretion through pharmacological activation. This study reveals an important physiological role for β cell O-GlcNAcylation in sensing and responding to obesity, with therapeutic implications for managing the relationship between type 2 diabetes and its most common risk factor.
Collapse
Affiliation(s)
- Amber Lockridge
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Seokwon Jo
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Eric Gustafson
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Niklas Damberg
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ramkumar Mohan
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Miranda Olson
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Juan E Abrahante
- Supercomputing Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Emilyn U Alejandro
- Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
50
|
Moslehi N, Mirmiran P, Marzbani R, Rezadoost H, Mirzaie M, Azizi F, Tehrani FR. Serum metabolomics study of women with different annual decline rates of anti-Müllerian hormone: an untargeted gas chromatography-mass spectrometry-based study. Hum Reprod 2021; 36:721-733. [PMID: 33320198 DOI: 10.1093/humrep/deaa279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/02/2020] [Indexed: 01/09/2023] Open
Abstract
STUDY QUESTION Which metabolites are associated with varying rates of ovarian aging, measured as annual decline rates of anti-Müllerian hormone (AMH) concentrations? SUMMARY ANSWER Higher serum concentrations of metabolites of phosphate, N-acetyl-d-glucosamine, branched chained amino acids (BCAAs), proline, urea and pyroglutamic acid were associated with higher odds of fast annual decline rate of AMH. WHAT IS KNOWN ALREADY Age-related rate of ovarian follicular loss varies among women, and the factors underlying such inter-individual variations are mainly unknown. The rate of ovarian aging is clinically important due to its effects on both reproduction and health of women. Metabolomics, a global investigation of metabolites in biological samples, provides an opportunity to study metabolites or metabolic pathways in relation to a physiological/pathophysiological condition. To date, no metabolomics study has been conducted regarding the differences in the rates of ovarian follicular loss. STUDY DESIGN, SIZE, DURATION This prospective study was conducted on 186 reproductive-aged women with regular menstrual cycles and history of natural fertility, randomly selected using random case selection option in SPSS from the Tehran Lipid and Glucose Study. PARTICIPANTS/MATERIALS, SETTING, METHODS AMH concentrations were measured at baseline (1999-2001) and the fifth follow-up examination (2014-2017), after a median follow-up of 16 years, by immunoassay using Gen II kit. The annual decline rate of AMH was calculated by dividing the AMH decline rate by the follow-up duration (percent/year). The women were categorized based on the tertiles of the annual decline rates. Untargeted metabolomics analysis of the fasting-serum samples collected during the second follow-up examination cycle (2005-2008) was performed using gas chromatography-mass spectrometry. A combination of univariate and multivariate approaches was used to investigate the associations between metabolites and the annual decline rates of AMH. MAIN RESULTS AND THE ROLE OF CHANCE After adjusting the baseline values of age, AMH and BMI, 29 metabolites were positively correlated with the annual AMH decline rates. The comparisons among the tertiles of the annual decline rate of AMH revealed an increase in the relative abundance of 15 metabolites in the women with a fast decline (tertile 3), compared to those with a slow decline (tertile 1). There was no distinct separation between women with slow and fast decline rates while considering 41 metabolites simultaneously using the principal component analysis and the partial least-squares discriminant analysis models. The odds of fast AMH decline was increased with higher serum metabolites of phosphate, N-acetyl-d-glucosamine, BCAAs, proline, urea and pyroglutamic acid. Amino sugar and nucleotide sugar metabolism, BCAAs metabolism and aminoacyl tRNA biosynthesis were among the most significant pathways associated with the fast decline rate of AMH. LIMITATIONS, REASONS FOR CAUTION Estimating the annual decline rates of AMH using the only two measures of AMH is the main limitation of the study which assumes a linear fixed reduction in AMH during the study. Since using the two-time points did not account for the variability in the decline rate of AMH, the annual decline rates estimated in this study may not accurately show the trend of the reduction in AMH. In addition, despite the longitudinal nature of the study and statistical adjustment of the participants' ages, it is difficult to distinguish the AMH-related metabolites observed in this study can accelerate ovarian aging or they are reflections of different rates of the aging process. WIDER IMPLICATIONS OF THE FINDINGS Some metabolite features related to the decline rates of AMH have been suggested in this study; further prospective studies with multiple measurements of AMH are needed to confirm the findings of this study and to better understand the molecular process underlying variations in ovarian aging. STUDY FUNDING/COMPETING INTEREST(S) This study, as a part of PhD thesis of Ms Nazanin Moslehi, was supported by Shahid Beheshti University of Medical Sciences (10522-4). There were no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Nazanin Moslehi
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Marzbani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Hassan Rezadoost
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Mehdi Mirzaie
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|