51
|
Gellai R, Hodrea J, Lenart L, Hosszu A, Koszegi S, Balogh D, Ver A, Banki NF, Fulop N, Molnar A, Wagner L, Vannay A, Szabo AJ, Fekete A. Role of O-linked N-acetylglucosamine modification in diabetic nephropathy. Am J Physiol Renal Physiol 2016; 311:F1172-F1181. [PMID: 27029430 DOI: 10.1152/ajprenal.00545.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/21/2016] [Indexed: 12/15/2022] Open
Abstract
Increased O-linked β-N-acetylglucosamine glycosylation (O-GlcNAcylation) is a known contributor to diabetes; however, its relevance in diabetic nephropathy (DN) is poorly elucidated. Here, we studied the process and enzymes of O-GlcNAcylation with a special emphasis on Akt-endothelial nitric oxide synthase (eNOS) and heat shock protein (HSP)72 signaling. Since tubular injury is the prominent site of DN, the effect of hyperglycemia was first measured in proximal tubular (HK2) cells cultured in high glucose. In vivo O-GlcNAcylation and protein levels of O-GlcNAc transferase (OGT), O-GlcNAcase (OGA), phosphorylated (p)Akt/Akt, peNOS/eNOS, and HSP72 were assessed in the kidney cortex of streptozotocin-induced diabetic rats. The effects of various renin-angiotensin-aldosterone system (RAAS) inhibitors were also evaluated. In proximal tubular cells, hyperglycemia-induced OGT expression led to increased O-GlcNAcylation, which was followed by a compensatory increase of OGA. In parallel, peNOS and pAkt levels decreased, whereas HSP72 increased. In diabetic rats, elevated O-GlcNAcylation was accompanied by decreased OGT and OGA. RAAS inhibitors ameliorated diabetes-induced kidney damage and prevented the elevation of O-GlcNAcylation and the decrement of pAkt, peNOS, and HSP72. In conclusion, hyperglycemia-induced elevation of O-GlcNAcylation contributes to the progression of DN via inhibition of Akt/eNOS phosphorylation and HSP72 induction. RAAS blockers successfully inhibit this process, suggesting a novel pathomechanism of their renoprotective action in the treatment of DN.
Collapse
Affiliation(s)
- Renata Gellai
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Judit Hodrea
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Lilla Lenart
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Adam Hosszu
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Sandor Koszegi
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Dora Balogh
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Agota Ver
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Nora F Banki
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Norbert Fulop
- Teaching Hospital Mór Kaposi, Kaposvar, Hungary; and
| | - Agnes Molnar
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Laszlo Wagner
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Adam Vannay
- MTA-SE Pediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Attila J Szabo
- MTA-SE Pediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Andrea Fekete
- MTA-SE "Lendulet" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary; .,First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| |
Collapse
|
52
|
Peterson SB, Hart GW. New insights: A role for O-GlcNAcylation in diabetic complications. Crit Rev Biochem Mol Biol 2016; 51:150-61. [DOI: 10.3109/10409238.2015.1135102] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
53
|
Makino A, Dai A, Han Y, Youssef KD, Wang W, Donthamsetty R, Scott BT, Wang H, Dillmann WH. O-GlcNAcase overexpression reverses coronary endothelial cell dysfunction in type 1 diabetic mice. Am J Physiol Cell Physiol 2015; 309:C593-9. [PMID: 26269457 PMCID: PMC4628934 DOI: 10.1152/ajpcell.00069.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the primary cause of morbidity and mortality in diabetes, and endothelial dysfunction is commonly seen in these patients. Increased O-linked N-acetylglucosamine (O-GlcNAc) protein modification is one of the central pathogenic features of diabetes. Modification of proteins by O-GlcNAc (O-GlcNAcylation) is regulated by two key enzymes: β-N-acetylglucosaminidase [O-GlcNAcase (OGA)], which catalyzes the reduction of protein O-GlcNAcylation, and O-GlcNAc transferase (OGT), which induces O-GlcNAcylation. However, it is not known whether reducing O-GlcNAcylation can improve endothelial dysfunction in diabetes. To examine the effect of endothelium-specific OGA overexpression on protein O-GlcNAcylation and coronary endothelial function in diabetic mice, we generated tetracycline-inducible, endothelium-specific OGA transgenic mice, and induced OGA by doxycycline administration in streptozotocin-induced type 1 diabetic mice. OGA protein expression was significantly decreased in mouse coronary endothelial cells (MCECs) isolated from diabetic mice compared with control MCECs, whereas OGT protein level was markedly increased. The level of protein O-GlcNAcylation was increased in diabetic compared with control mice, and OGA overexpression significantly decreased the level of protein O-GlcNAcylation in MCECs from diabetic mice. Capillary density in the left ventricle and endothelium-dependent relaxation in coronary arteries were significantly decreased in diabetes, while OGA overexpression increased capillary density to the control level and restored endothelium-dependent relaxation without changing endothelium-independent relaxation. We found that connexin 40 could be the potential target of O-GlcNAcylation that regulates the endothelial functions in diabetes. These data suggest that OGA overexpression in endothelial cells improves endothelial function and may have a beneficial effect on coronary vascular complications in diabetes.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Cells, Cultured
- Connexins/metabolism
- Coronary Artery Disease/enzymology
- Coronary Artery Disease/genetics
- Coronary Artery Disease/physiopathology
- Coronary Vessels/drug effects
- Coronary Vessels/enzymology
- Coronary Vessels/physiopathology
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/physiopathology
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/physiopathology
- Endothelial Cells/drug effects
- Endothelial Cells/enzymology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Enzyme Induction
- Enzyme Inhibitors/pharmacology
- Glycosylation
- Histone Acetyltransferases/antagonists & inhibitors
- Histone Acetyltransferases/biosynthesis
- Histone Acetyltransferases/genetics
- Humans
- Hyaluronoglucosaminidase/antagonists & inhibitors
- Hyaluronoglucosaminidase/biosynthesis
- Hyaluronoglucosaminidase/genetics
- Male
- Mice, Transgenic
- N-Acetylglucosaminyltransferases/metabolism
- Neovascularization, Physiologic
- Protein Processing, Post-Translational
- Signal Transduction
- Vasodilation
- beta-N-Acetylhexosaminidases/antagonists & inhibitors
- beta-N-Acetylhexosaminidases/biosynthesis
- beta-N-Acetylhexosaminidases/genetics
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Ayako Makino
- Department of Physiology, University of Arizona, Tucson, Arizona; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Department of Medicine, University of California, San Diego, La Jolla, California
| | - Anzhi Dai
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ying Han
- Department of Physiology, University of Arizona, Tucson, Arizona; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Katia D Youssef
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Weihua Wang
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Reshma Donthamsetty
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Hong Wang
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
54
|
Issad T. O-GlcNAcylation of connexin 40: a sweet connection between diabetes and endothelial cell dysfunction? Focus on “O-GlcNAcase overexpression reverses coronary endothelial cell dysfunction in type 1 diabetic mice”. Am J Physiol Cell Physiol 2015; 309:C590-2. [DOI: 10.1152/ajpcell.00260.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Tarik Issad
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; and Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
55
|
CHAIYAWAT PARUNYA, CHOKCHAICHAMNANKIT DARANEE, LIRDPRAPAMONGKOL KRIENGSAK, SRISOMSAP CHANTRAGAN, SVASTI JISNUSON, CHAMPATTANACHAI VORARATT. Alteration of O-GlcNAcylation affects serine phosphorylation and regulates gene expression and activity of pyruvate kinase M2 in colorectal cancer cells. Oncol Rep 2015; 34:1933-42. [DOI: 10.3892/or.2015.4178] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/27/2015] [Indexed: 11/05/2022] Open
|
56
|
|
57
|
Goveia J, Stapor P, Carmeliet P. Principles of targeting endothelial cell metabolism to treat angiogenesis and endothelial cell dysfunction in disease. EMBO Mol Med 2015; 6:1105-20. [PMID: 25063693 PMCID: PMC4197858 DOI: 10.15252/emmm.201404156] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The endothelium is the orchestral conductor of blood vessel function. Pathological blood vessel formation (a process termed pathological angiogenesis) or the inability of endothelial cells (ECs) to perform their physiological function (a condition known as EC dysfunction) are defining features of various diseases. Therapeutic intervention to inhibit aberrant angiogenesis or ameliorate EC dysfunction could be beneficial in diseases such as cancer and cardiovascular disease, respectively, but current strategies have limited efficacy. Based on recent findings that pathological angiogenesis and EC dysfunction are accompanied by EC-specific metabolic alterations, targeting EC metabolism is emerging as a novel therapeutic strategy. Here, we review recent progress in our understanding of how EC metabolism is altered in disease and discuss potential metabolic targets and strategies to reverse EC dysfunction and inhibit pathological angiogenesis.
Collapse
Affiliation(s)
- Jermaine Goveia
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center VIB, Leuven, Belgium
| | - Peter Stapor
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center VIB, Leuven, Belgium
| |
Collapse
|
58
|
|
59
|
Zheng Y, Cao X, Wen J, Yang H, Luo K, Liu Q, Huang Q, Chen J, Fu J. Smoking affects treatment outcome in patients with resected esophageal squamous cell carcinoma who received chemotherapy. PLoS One 2015; 10:e0123246. [PMID: 25874561 PMCID: PMC4395356 DOI: 10.1371/journal.pone.0123246] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/02/2015] [Indexed: 01/25/2023] Open
Abstract
Background Cigarette smoking is reported to decrease survival and induce chemotherapy resistance in patients with various cancers. However, the impact of cigarette smoking on patients with esophageal squamous cell carcinoma (ESCC) remains unknown. Methods A total of 1,084 ESCC patients were retrospectively enrolled from a southern Chinese institution. Patients were divided into two groups according to their treatment modalities: the SC group (surgery with chemotherapy) (n = 306) and the S group (surgery without chemotherapy) (n = 778). Smoking status was quantified as smoking history (non-smoker, ex-smoker, and current smoker) and cumulative smoking (0, between 0 and 20, and greater than 20 pack-years). The association between cigarette smoking and overall survival (OS) was evaluated using the Kaplan-Meier method and univariate/multivariate regression analysis. Results Among 1,084 patients, 702 (64.8%) reported a cigarette smoking history, and the 5-year OS for non-smokers and smokers was 45.8% and 37.3%, respectively. In the SC group, compared with non-smoker, the adjusted HRs of ex-smoker and current smoker were 1.540 (95% CI, 1.1–2.2) and 2.110 (95% CI, 1.4–3.1), respectively; there is a correlative trend of decreased OS with increased cigarette smoking (Ptrend = 0.001). These associations were insignificant in the S group. In subgroup analysis of the SC group, the lower OS conferred by smoking was not significantly modified by age, gender, body mass index, alcohol drinking, or chemotherapy method (chemotherapy and chemoradiotherapy). Conclusion Our results suggest that smoking may affect treatment outcome in patients with resected ESCC who received chemotherapy.
Collapse
Affiliation(s)
- Yuzhen Zheng
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Xun Cao
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
| | - Jing Wen
- Department of Experimental Research, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Hong Yang
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Kongjia Luo
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Qianwen Liu
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Qingyuan Huang
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Junying Chen
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
| | - Jianhua Fu
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation, Guangzhou, Guangdong, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, P.R. China
- * E-mail:
| |
Collapse
|
60
|
Abstract
Unlike the complex glycans decorating the cell surface, the O-linked β-N-acetyl glucosamine (O-GlcNAc) modification is a simple intracellular Ser/Thr-linked monosaccharide that is important for disease-relevant signaling and enzyme regulation. O-GlcNAcylation requires uridine diphosphate-GlcNAc, a precursor responsive to nutrient status and other environmental cues. Alternative splicing of the genes encoding the O-GlcNAc cycling enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) yields isoforms targeted to discrete sites in the nucleus, cytoplasm, and mitochondria. OGT and OGA also partner with cellular effectors and act in tandem with other posttranslational modifications. The enzymes of O-GlcNAc cycling act preferentially on intrinsically disordered domains of target proteins impacting transcription, metabolism, apoptosis, organelle biogenesis, and transport.
Collapse
|
61
|
Verdegem D, Moens S, Stapor P, Carmeliet P. Endothelial cell metabolism: parallels and divergences with cancer cell metabolism. Cancer Metab 2014; 2:19. [PMID: 25250177 PMCID: PMC4171726 DOI: 10.1186/2049-3002-2-19] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/14/2014] [Indexed: 02/08/2023] Open
Abstract
The stromal vasculature in tumors is a vital conduit of nutrients and oxygen for cancer cells. To date, the vast majority of studies have focused on unraveling the genetic basis of vessel sprouting (also termed angiogenesis). In contrast to the widely studied changes in cancer cell metabolism, insight in the metabolic regulation of angiogenesis is only just emerging. These studies show that metabolic pathways in endothelial cells (ECs) importantly regulate angiogenesis in conjunction with genetic signals. In this review, we will highlight these emerging insights in EC metabolism and discuss them in perspective of cancer cell metabolism. While it is generally assumed that cancer cells have unique metabolic adaptations, not shared by healthy non-transformed cells, we will discuss parallels and highlight differences between endothelial and cancer cell metabolism and consider possible novel therapeutic opportunities arising from targeting both cancer and endothelial cells.
Collapse
Affiliation(s)
- Dries Verdegem
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Stijn Moens
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Peter Stapor
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| |
Collapse
|
62
|
Stapor P, Wang X, Goveia J, Moens S, Carmeliet P. Angiogenesis revisited - role and therapeutic potential of targeting endothelial metabolism. J Cell Sci 2014; 127:4331-41. [PMID: 25179598 DOI: 10.1242/jcs.153908] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Clinically approved therapies that target angiogenesis in tumors and ocular diseases focus on controlling pro-angiogenic growth factors in order to reduce aberrant microvascular growth. Although research on angiogenesis has revealed key mechanisms that regulate tissue vascularization, therapeutic success has been limited owing to insufficient efficacy, refractoriness and tumor resistance. Emerging concepts suggest that, in addition to growth factors, vascular metabolism also regulates angiogenesis and is a viable target for manipulating the microvasculature. Recent studies show that endothelial cells rely on glycolysis for ATP production, and that the key glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) regulates angiogenesis by controlling the balance of tip versus stalk cells. As endothelial cells acquire a tip cell phenotype, they increase glycolytic production of ATP for sprouting. Furthermore, pharmacological blockade of PFKFB3 causes a transient, partial reduction in glycolysis, and reduces pathological angiogenesis with minimal systemic harm. Although further assessment of endothelial cell metabolism is necessary, these results represent a paradigm shift in anti-angiogenic therapy from targeting angiogenic factors to focusing on vascular metabolism, warranting research on the metabolic pathways that govern angiogenesis.
Collapse
Affiliation(s)
- Peter Stapor
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Xingwu Wang
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Stijn Moens
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
63
|
Abstract
Ocular neovascularization can affect almost all the tissues of the eye: the cornea, the iris, the retina, and the choroid. Pathological neovascularization is the underlying cause of vision loss in common ocular conditions such as diabetic retinopathy, retinopathy of prematurity and age-related macular neovascularization. Glycosylation is the most common covalent posttranslational modification of proteins in mammalian cells. A growing body of evidence demonstrates that glycosylation influences the process of angiogenesis and impacts activation, proliferation, and migration of endothelial cells as well as the interaction of angiogenic endothelial cells with other cell types necessary to form blood vessels. Recent studies have provided evidence that members of the galectin class of β-galactoside-binding proteins modulate angiogenesis by novel carbohydrate-based recognition systems involving interactions between glycans of angiogenic cell surface receptors and galectins. This review discusses the significance of glycosylation and the role of galectins in the pathogenesis of ocular neovascularization.
Collapse
Affiliation(s)
- Anna I Markowska
- Departments of Ophthalmology and Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA Ymir Genomics LLC, Cambridge, MA 02139, USA
| | - Zhiyi Cao
- Departments of Ophthalmology and Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA New England Eye Center, Boston, MA 02111, USA
| | - Noorjahan Panjwani
- Departments of Ophthalmology and Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
64
|
Ghesquière B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature 2014; 511:167-76. [DOI: 10.1038/nature13312] [Citation(s) in RCA: 318] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/08/2014] [Indexed: 12/11/2022]
|
65
|
Baudoin L, Issad T. O-GlcNAcylation and Inflammation: A Vast Territory to Explore. Front Endocrinol (Lausanne) 2014; 5:235. [PMID: 25620956 PMCID: PMC4288382 DOI: 10.3389/fendo.2014.00235] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 12/18/2014] [Indexed: 01/04/2023] Open
Abstract
O-GlcNAcylation is a reversible post-translational modification that regulates the activities of cytosolic and nuclear proteins according to glucose availability. This modification appears to participate in several hyperglycemia-associated complications. An important feature of metabolic diseases such as diabetes and obesity is the presence of a low-grade chronic inflammation that causes numerous complications. Hyperglycemia associated with the metabolic syndrome is known to promote inflammatory processes through different mechanisms including oxidative stress and abnormally elevated protein O-GlcNAcylation. However, the role of O-GlcNAcylation on inflammation remains contradictory. O-GlcNAcylation associated with hyperglycemia has been shown to increase nuclear factor κB (NFκB) transcriptional activity through different mechanisms. This could contribute in inflammation-associated diabetic complications. However, in other conditions such as acute vascular injury, O-linked N-acetyl glucosamine (O-GlcNAc) also exerts anti-inflammatory effects via inhibition of the NFκB pathway, suggesting a complex regulation of inflammation by O-GlcNAc. Moreover, whereas macrophages and monocytes exposed to high glucose for a long-term period developed a pro-inflammatory phenotype, the impact of O-GlcNAcylation in these cells remains unclear. A future challenge will be to clearly establish the role of O-GlcNAcylation in pro- and anti-inflammatory functions in macrophages.
Collapse
Affiliation(s)
- Léa Baudoin
- UMR8104, CNRS, Institut Cochin, Université Paris Descartes, Paris, France
- U1016, INSERM, Paris, France
| | - Tarik Issad
- UMR8104, CNRS, Institut Cochin, Université Paris Descartes, Paris, France
- U1016, INSERM, Paris, France
- *Correspondence: Tarik Issad, Department of Endocrinology, Metabolism and Diabetes, Institute Cochin, 22 rue Méchain, Paris 75014, France e-mail:
| |
Collapse
|
66
|
Abstract
Endothelial cells (ECs) are quiescent for years but can plastically switch to angiogenesis. Vascular sprouting relies on the coordinated activity of migrating tip cells at the forefront and proliferating stalk cells that elongate the sprout. Past studies have identified genetic signals that control vascular branching. Prominent are VEGF, activating tip cells, and Notch, which stimulates stalk cells. After the branch is formed and perfused, ECs become quiescent phalanx cells. Now, emerging evidence has accumulated indicating that ECs not only adapt their metabolism when switching from quiescence to sprouting but also that metabolism regulates vascular sprouting in parallel to the control by genetic signals.
Collapse
Affiliation(s)
- Katrien De Bock
- Department of Oncology, University of Leuven, Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Leuven 3000, Belgium; VIB, Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Leuven 3000, Belgium
| | | | | |
Collapse
|
67
|
Gurel Z, Sieg KM, Shallow KD, Sorenson CM, Sheibani N. Retinal O-linked N-acetylglucosamine protein modifications: implications for postnatal retinal vascularization and the pathogenesis of diabetic retinopathy. Mol Vis 2013; 19:1047-59. [PMID: 23734074 PMCID: PMC3668662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 05/18/2013] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Hyperglycemia activates several metabolic pathways, including the hexosamine biosynthetic pathway. Uridine diphosphate N-acetylglucosamine (GlcNAc) is the product of the hexosamine biosynthetic pathway and the substrate for O-linked GlcNAc (O-GlcNAc) modification. This modification affects a wide range of proteins by altering their activity, cellular localization, and/or protein interactions. However, the role O-GlcNAcylation may play in normal postnatal retinal vascular development and in the ocular complications of diabetes, including diabetic retinopathy, requires further investigation. METHODS The total levels of O-GlcNAc-modified proteins were evaluated by western blot analysis of lysates prepared from retinas obtained at different days during postnatal retinal vascularization and oxygen-induced ischemic retinopathy. Similar experiments were performed with retinal lysate prepared from diabetic Ins2(Akita/+) mice with different durations of diabetes and retinal vascular cells cultured under various glucose conditions. The localization of O-GlcNAc-modified proteins in the retinal vasculature was confirmed by immunofluorescence staining. The impact of altered O-GlcNAcylation on the migration of retinal vascular cells was determined using scratch wound and transwell migration assays. RESULTS We detected an increase in protein O-GlcNAcylation during mouse postnatal retinal vascularization and aging, in part through the regulation of the enzymes that control this modification. The study of the diabetic Ins2(Akita/+) mouse retina showed an increase in the O-GlcNAc modification of retinal proteins. We also observed an increase in retinal O-GlcNAcylated protein levels during the neovascularization phase of oxygen-induced ischemic retinopathy. Our fluorescence microscopy data confirmed that the alterations in retinal O-GlcNAcylation are similarly represented in the retinal vasculature and in retinal pericytes and endothelial cells. Particularly, the migration of retinal pericytes, but not retinal endothelial cells, was attenuated by increased O-GlcNAc modification. CONCLUSIONS The O-GlcNAc modification pattern changes during postnatal retinal vascular development and neovascularization, and its dysregulation under hyperglycemia and/or ischemia may contribute to the pathogenesis of the diabetic retinopathy and retinal neovascularization.
Collapse
Affiliation(s)
- Zafer Gurel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison,WI
| | - Kelsey M. Sieg
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison,WI
| | - Keegan D. Shallow
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison,WI
| | - Christine M. Sorenson
- Department Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison,WI,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison,WI
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison,WI,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison,WI
| |
Collapse
|
68
|
Ding F, Yu L, Wang M, Xu S, Xia Q, Fu G. O-GlcNAcylation involvement in high glucose-induced cardiac hypertrophy via ERK1/2 and cyclin D2. Amino Acids 2013; 45:339-49. [PMID: 23665912 DOI: 10.1007/s00726-013-1504-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 04/19/2013] [Indexed: 01/26/2023]
Abstract
Continuous hyperglycemia is considered to be the most significant pathogenesis of diabetic cardiomyopathy, which manifests as cardiac hypertrophy and subsequent heart failure. O-GlcNAcylation has attracted attention as a post-translational protein modification in the past decade. The role of O-GlcNAcylation in high glucose-induced cardiomyocyte hypertrophy remains unclear. We studied the effect of O-GlcNAcylation on neonatal rat cardiomyocytes that were exposed to high glucose and myocardium in diabetic rats induced by streptozocin. High glucose (30 mM) incubation induced a greater than twofold increase in cell size and increased hypertrophy marker gene expression accompanied by elevated O-GlcNAcylation protein levels. High glucose increased ERK1/2 but not p38 MAPK or JNK activity, and cyclin D2 expression was also increased. PUGNAc, an inhibitor of β-N-acetylglucosaminidase, enhanced O-GlcNAcylation and imitated the effects of high glucose. OGT siRNA and ERK1/2 inhibition with PD98059 treatment blunted the hypertrophic response and cyclin D2 upregulation. OGT inhibition also prevented ERK1/2 activation. We also observed concentric hypertrophy and similar changes of O-GlcNAcylation level, ERK1/2 activation and cyclin D2 expression in myocardium of diabetic rats induced by streptozocin. In conclusion, O-GlcNAcylation plays a role in high glucose-induced cardiac hypertrophy via ERK1/2 and cyclin D2.
Collapse
Affiliation(s)
- Fang Ding
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, China
| | | | | | | | | | | |
Collapse
|
69
|
Abstract
To maintain homeostasis under variable nutrient conditions, cells rapidly and robustly respond to fluctuations through adaptable signaling networks. Evidence suggests that the O-linked N-acetylglucosamine (O-GlcNAc) posttranslational modification of serine and threonine residues functions as a critical regulator of intracellular signaling cascades in response to nutrient changes. O-GlcNAc is a highly regulated, reversible modification poised to integrate metabolic signals and acts to influence many cellular processes, including cellular signaling, protein stability, and transcription. This review describes the role O-GlcNAc plays in governing both integrated cellular processes and the activity of individual proteins in response to nutrient levels. Moreover, we discuss the ways in which cellular changes in O-GlcNAc status may be linked to chronic diseases such as type 2 diabetes, neurodegeneration, and cancers, providing a unique window through which to identify and treat disease conditions.
Collapse
Affiliation(s)
- Michelle R. Bond
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; ,
| | - John A. Hanover
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; ,
| |
Collapse
|
70
|
Abstract
It is increasingly apparent that not only is a cure for the current worldwide diabetes epidemic required, but also for its major complications, affecting both small and large blood vessels. These complications occur in the majority of individuals with both type 1 and type 2 diabetes. Among the most prevalent microvascular complications are kidney disease, blindness, and amputations, with current therapies only slowing disease progression. Impaired kidney function, exhibited as a reduced glomerular filtration rate, is also a major risk factor for macrovascular complications, such as heart attacks and strokes. There have been a large number of new therapies tested in clinical trials for diabetic complications, with, in general, rather disappointing results. Indeed, it remains to be fully defined as to which pathways in diabetic complications are essentially protective rather than pathological, in terms of their effects on the underlying disease process. Furthermore, seemingly independent pathways are also showing significant interactions with each other to exacerbate pathology. Interestingly, some of these pathways may not only play key roles in complications but also in the development of diabetes per se. This review aims to comprehensively discuss the well validated, as well as putative mechanisms involved in the development of diabetic complications. In addition, new fields of research, which warrant further investigation as potential therapeutic targets of the future, will be highlighted.
Collapse
Affiliation(s)
- Josephine M Forbes
- Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | |
Collapse
|
71
|
Liu H, Yu S, Zhang H, Xu J. Angiogenesis impairment in diabetes: role of methylglyoxal-induced receptor for advanced glycation endproducts, autophagy and vascular endothelial growth factor receptor 2. PLoS One 2012; 7:e46720. [PMID: 23056421 PMCID: PMC3463541 DOI: 10.1371/journal.pone.0046720] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 09/07/2012] [Indexed: 12/20/2022] Open
Abstract
Diabetes impairs physiological angiogenesis by molecular mechanisms that are not fully understood. Methylglyoxal (MGO), a metabolite of glycolysis, is increased in patients with diabetes. This study defined the role of MGO in angiogenesis impairment and tested the mechanism in diabetic animals. Endothelial cells and mouse aortas were subjected to Western blot analysis of vascular endothelial growth factor receptor 2 (VEGFR2) protein levels and angiogenesis evaluation by endothelial cell tube formation/migration and aortic ring assays. Incubation with MGO reduced VEGFR2 protein, but not mRNA, levels in a time and dose dependent manner. Genetic knockdown of the receptor for advanced glycation endproducts (RAGE) attenuated the reduction of VEGFR2. Overexpression of Glyoxalase 1, the enzyme that detoxifies MGO, reduced the MGO-protein adducts and prevented VEGFR2 reduction. The VEGFR2 reduction was associated with impaired angiogenesis. Suppression of autophagy either by inhibitors or siRNA, but not of the proteasome and caspase, normalized both the VEGFR2 protein levels and angiogenesis. Conversely, induction of autophagy either by rapamycin or overexpression of LC3 and Beclin-1 reduced VEGFR2 and angiogenesis. MGO increased endothelial LC3B and Beclin-1, markers of autophagy, which were accompanied by an increase of both autophagic flux (LC3 punctae) and co-immunoprecipitation of VEGFR2 with LC3. Pharmacological or genetic suppression of peroxynitrite (ONOO(-)) generation not only blocked the autophagy but also reversed the reduction of VEGFR2 and angiogenesis. Like MGO-treated aortas from normglycemic C57BL/6J mice, aortas from diabetic db/db and Akita mice presented reductions of angiogenesis or VEGFR2. Administration of either autophagy inhibitor ex vivo or superoxide scavenger in vivo abolished the reductions. Taken together, MGO reduces endothelial angiogenesis through RAGE-mediated, ONOO(-)dependent and autophagy-induced VEGFR2 degradation, which may represent a new mechanism for diabetic angiogenesis impairment.
Collapse
Affiliation(s)
- Hongtao Liu
- Section of Endocrinology, Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shujie Yu
- Section of Endocrinology, Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Hua Zhang
- Section of Endocrinology, Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jian Xu
- Section of Endocrinology, Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
72
|
Osaki T, Azuma K, Kurozumi S, Takamori Y, Tsuka T, Imagawa T, Okamoto Y, Minami S. Metabolomic analyses of blood plasma after oral administration of D-glucosamine hydrochloride to dogs. Mar Drugs 2012; 10:1873-1882. [PMID: 23015778 PMCID: PMC3447342 DOI: 10.3390/md10081873] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 07/02/2012] [Accepted: 08/15/2012] [Indexed: 11/16/2022] Open
Abstract
D-Glucosamine hydrochloride (GlcN∙HCl) is an endogenous amino monosaccharide synthesized from glucose that is useful in the treatment of joint diseases in both humans and animals. The aim of this study was to examine amino acid metabolism in dogs after oral administration of GlcN∙HCl. Accelerated fumarate respiration and elevated plasma levels of lactic acid and alanine were observed after administration. These results suggest that oral administration of GlcN∙HCl induces anaerobic respiration and starvation in cells, and we hypothesize that these conditions promote cartilage regeneration. Further studies are required to evaluate the expression of transforming growth factor-beta (TGF-β).
Collapse
Affiliation(s)
- Tomohiro Osaki
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan; (K.A.); (T.T.); (T.I.); (Y.O.); (S.M.)
- Author to whom correspondence should be addressed; ; Tel./Fax: +81-857-31-5434
| | - Kazuo Azuma
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan; (K.A.); (T.T.); (T.I.); (Y.O.); (S.M.)
| | - Seiji Kurozumi
- Koyo Chemical Co. Ltd., 3-11-15 Iidabashi, Chiyodaku, Tokyo 102-0072, Japan; (S.K.); (Y.T.)
| | - Yoshimori Takamori
- Koyo Chemical Co. Ltd., 3-11-15 Iidabashi, Chiyodaku, Tokyo 102-0072, Japan; (S.K.); (Y.T.)
| | - Takeshi Tsuka
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan; (K.A.); (T.T.); (T.I.); (Y.O.); (S.M.)
| | - Tomohiro Imagawa
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan; (K.A.); (T.T.); (T.I.); (Y.O.); (S.M.)
| | - Yoshiharu Okamoto
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan; (K.A.); (T.T.); (T.I.); (Y.O.); (S.M.)
| | - Saburo Minami
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan; (K.A.); (T.T.); (T.I.); (Y.O.); (S.M.)
| |
Collapse
|
73
|
Diverse regulation of AKT and GSK-3β by O-GlcNAcylation in various types of cells. FEBS Lett 2012; 586:2443-50. [PMID: 22687243 DOI: 10.1016/j.febslet.2012.05.063] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 05/23/2012] [Accepted: 05/31/2012] [Indexed: 01/01/2023]
Abstract
Protein kinase B (AKT) and glycogen synthase kinase-3β (GSK-3β) are major components of insulin-AKT signaling that plays crucial roles in various types of tissue. Recent studies found that these two kinases are modified posttranslationally by O-GlcNAcylation. Here, we demonstrate that O-GlcNAcylation regulated phosphorylation/activation of AKT and GSK-3β in different manners in kidney HEK-293FT cells, but did not affect these two kinases in hepatic HepG2 cells. In neuronal cells, O-GlcNAcylation regulated phosphorylation of AKT negatively, but had no effect on GSK-3β. These results suggest protein-specific and cell type-specific regulation of AKT and GSK-3β by O-GlcNAcylation. Therefore, studies on the roles of AKT and GSK-3β O-GlcNAcylation should be done in a tissue- and cell type-specific manner.
Collapse
|
74
|
Dias WB, Cheung WD, Hart GW. O-GlcNAcylation of kinases. Biochem Biophys Res Commun 2012; 422:224-8. [PMID: 22564745 PMCID: PMC3387735 DOI: 10.1016/j.bbrc.2012.04.124] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 04/22/2012] [Indexed: 01/17/2023]
Abstract
Recent evidence indicates that site-specific crosstalk between O-GlcNAcylation and phosphorylation and the O-GlcNAcylation of kinases play an important role in regulating cell signaling. However, relatively few kinases have been analyzed for O-GlcNAcylation. Here, we identify additional kinases that are substrates for O-GlcNAcylation using an in vitro OGT assay on a functional kinase array. Forty-two kinases were O-GlcNAcylated in vitro, representing 39% of the kinases on the array. In addition, we confirmed the in vivo O-GlcNAcylation of three identified kinases. Our results suggest that O-GlcNAcylation may directly regulate a substantial number of kinases and illustrates the increasingly complex relationship between O-GlcNAcylation and phosphorylation in cellular signaling.
Collapse
Affiliation(s)
- Wagner B Dias
- Department of Biological Chemistry, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
75
|
Yu Y, Zhang L, Li X, Run X, Liang Z, Li Y, Liu Y, Lee MH, Grundke-Iqbal I, Iqbal K, Vocadlo DJ, Liu F, Gong CX. Differential effects of an O-GlcNAcase inhibitor on tau phosphorylation. PLoS One 2012; 7:e35277. [PMID: 22536363 PMCID: PMC3334936 DOI: 10.1371/journal.pone.0035277] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 03/15/2012] [Indexed: 11/18/2022] Open
Abstract
Abnormal hyperphosphorylation of microtubule-associated protein tau plays a crucial role in neurodegeneration in Alzheimer's disease (AD). The aggregation of hyperphosphorylated tau into neurofibrillary tangles is also a hallmark brain lesion of AD. Tau phosphorylation is regulated by tau kinases, tau phosphatases, and O-GlcNAcylation, a posttranslational modification of proteins on the serine or threonine residues with β-N-acetylglucosamine (GlcNAc). O-GlcNAcylation is dynamically regulated by O-GlcNAc transferase, the enzyme catalyzing the transfer of GlcNAc to proteins, and N-acetylglucosaminidase (OGA), the enzyme catalyzing the removal of GlcNAc from proteins. Thiamet-G is a recently synthesized potent OGA inhibitor, and initial studies suggest it can influence O-GlcNAc levels in the brain, allowing OGA inhibition to be a potential route to altering disease progression in AD. In this study, we injected thiamet-G into the lateral ventricle of mice to increase O-GlcNAcylation of proteins and investigated the resulting effects on site-specific tau phosphorylation. We found that acute thiamet-G treatment led to a decrease in tau phosphorylation at Thr181, Thr212, Ser214, Ser262/Ser356, Ser404 and Ser409, and an increase in tau phosphorylation at Ser199, Ser202, Ser396 and Ser422 in the mouse brain. Investigation of the major tau kinases showed that acute delivery of a high dose of thiamet-G into the brain also led to a marked activation of glycogen synthase kinase-3β (GSK-3β), possibly as a consequence of down-regulation of its upstream regulating kinase, AKT. However, the elevation of tau phosphorylation at the sites above was not observed and GSK-3β was not activated in cultured adult hippocampal progenitor cells or in PC12 cells after thiamet-G treatment. These results suggest that acute high-dose thiamet-G injection can not only directly antagonize tau phosphorylation, but also stimulate GSK-3β activity, with the downstream consequence being site-specific, bi-directional regulation of tau phosphorylation in the mammalian brain.
Collapse
Affiliation(s)
- Yang Yu
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Lan Zhang
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Xiaojing Li
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Xiaoqin Run
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Zhihou Liang
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Yi Li
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Ying Liu
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Moon H. Lee
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Inge Grundke-Iqbal
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Khalid Iqbal
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - David J. Vocadlo
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Fei Liu
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Cheng-Xin Gong
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| |
Collapse
|
76
|
Zachara NE. The roles of O-linked β-N-acetylglucosamine in cardiovascular physiology and disease. Am J Physiol Heart Circ Physiol 2012; 302:H1905-18. [PMID: 22287582 DOI: 10.1152/ajpheart.00445.2011] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
More than 1,000 proteins of the nucleus, cytoplasm, and mitochondria are dynamically modified by O-linked β-N-acetylglucosamine (O-GlcNAc), an essential post-translational modification of metazoans. O-GlcNAc, which modifies Ser/Thr residues, is thought to regulate protein function in a manner analogous to protein phosphorylation and, on a subset of proteins, appears to have a reciprocal relationship with phosphorylation. Like phosphorylation, O-GlcNAc levels change dynamically in response to numerous signals including hyperglycemia and cellular injury. Recent data suggests that O-GlcNAc appears to be a key regulator of the cellular stress response, the augmentation of which is protective in models of acute vascular injury, trauma hemorrhage, and ischemia-reperfusion injury. In contrast to these studies, O-GlcNAc has also been implicated in the development of hypertension and type II diabetes, leading to vascular and cardiac dysfunction. Here we summarize the current understanding of the roles of O-GlcNAc in the heart and vasculature.
Collapse
Affiliation(s)
- Natasha E Zachara
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
77
|
Hart GW, Slawson C, Ramirez-Correa G, Lagerlof O. Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. Annu Rev Biochem 2011; 80:825-58. [PMID: 21391816 DOI: 10.1146/annurev-biochem-060608-102511] [Citation(s) in RCA: 1030] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
O-GlcNAcylation is the addition of β-D-N-acetylglucosamine to serine or threonine residues of nuclear and cytoplasmic proteins. O-linked N-acetylglucosamine (O-GlcNAc) was not discovered until the early 1980s and still remains difficult to detect and quantify. Nonetheless, O-GlcNAc is highly abundant and cycles on proteins with a timescale similar to protein phosphorylation. O-GlcNAc occurs in organisms ranging from some bacteria to protozoans and metazoans, including plants and nematodes up the evolutionary tree to man. O-GlcNAcylation is mostly on nuclear proteins, but it occurs in all intracellular compartments, including mitochondria. Recent glycomic analyses have shown that O-GlcNAcylation has surprisingly extensive cross talk with phosphorylation, where it serves as a nutrient/stress sensor to modulate signaling, transcription, and cytoskeletal functions. Abnormal amounts of O-GlcNAcylation underlie the etiology of insulin resistance and glucose toxicity in diabetes, and this type of modification plays a direct role in neurodegenerative disease. Many oncogenic proteins and tumor suppressor proteins are also regulated by O-GlcNAcylation. Current data justify extensive efforts toward a better understanding of this invisible, yet abundant, modification. As tools for the study of O-GlcNAc become more facile and available, exponential growth in this area of research will eventually take place.
Collapse
Affiliation(s)
- Gerald W Hart
- Departments of Biological Chemistry and Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205
| | | | | | | |
Collapse
|
78
|
Soesanto Y, Luo B, Parker G, Jones D, Cooksey RC, McClain DA. Pleiotropic and age-dependent effects of decreased protein modification by O-linked N-acetylglucosamine on pancreatic β-cell function and vascularization. J Biol Chem 2011; 286:26118-26. [PMID: 21622566 DOI: 10.1074/jbc.m111.249508] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The hexosamine biosynthesis pathway (HBP) regulates the post-translational modification of nuclear and cytoplasmic protein by O-linked N-acetylglucosamine (O-GlcNAc). Numerous studies have demonstrated increased flux through this pathway contributes to the development of β-cell dysfunction. The effect of decreased O-GlcNAc on the maintenance of normal β-cell function, however, is not well understood. We studied transgenic mice that over express β-N-acetylglucosaminidase (O-GlcNAcase), an enzyme that catalyzes the removal of O-GlcNAc from proteins, in the pancreatic β-cell under control of the rat insulin promoter. 3-4-Month-old O-GlcNAcase transgenic mice have higher glucose excursions with a concomitant decrease in circulating insulin levels, insulin mRNA levels, and total islet insulin content. In older (8-9-month-old) O-GlcNAcase transgenic mice glucose tolerance is no longer impaired. This is associated with increased serum insulin, islet insulin content, and insulin mRNA in the O-GlcNAcase transgenic mice. These improvements in β-cell function with aging are associated with increased angiogenesis and increased VEGF expression, with parallel increases in activation of Akt and expression of PGC1α. The biphasic effects as a function of age are consistent with published observations of mice with increased O-GlcNAc in islets and demonstrate that O-GlcNAc signaling exerts multiple effects on both insulin secretion and islet survival.
Collapse
Affiliation(s)
- Yudi Soesanto
- Departments of Biochemistry, University of Utah School ofMedicine, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | |
Collapse
|
79
|
Acute regulation of cardiac metabolism by the hexosamine biosynthesis pathway and protein O-GlcNAcylation. PLoS One 2011; 6:e18417. [PMID: 21494549 PMCID: PMC3073970 DOI: 10.1371/journal.pone.0018417] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 03/07/2011] [Indexed: 02/07/2023] Open
Abstract
Objective The hexosamine biosynthesis pathway (HBP) flux and protein O-linked N-acetyl-glucosamine (O-GlcNAc) levels have been implicated in mediating the adverse effects of diabetes in the cardiovascular system. Activation of these pathways with glucosamine has been shown to mimic some of the diabetes-induced functional and structural changes in the heart; however, the effect on cardiac metabolism is not known. Therefore, the primary goal of this study was to determine the effects of glucosamine on cardiac substrate utilization. Methods Isolated rat hearts were perfused with glucosamine (0–10 mM) to increase HBP flux under normoxic conditions. Metabolic fluxes were determined by 13C-NMR isotopomer analysis; UDP-GlcNAc a precursor of O-GlcNAc synthesis was assessed by HPLC and immunoblot analysis was used to determine O-GlcNAc levels, phospho- and total levels of AMPK and ACC, and membrane levels of FAT/CD36. Results Glucosamine caused a dose dependent increase in both UDP-GlcNAc and O-GlcNAc levels, which was associated with a significant increase in palmitate oxidation with a concomitant decrease in lactate and pyruvate oxidation. There was no effect of glucosamine on AMPK or ACC phosphorylation; however, membrane levels of the fatty acid transport protein FAT/CD36 were increased and preliminary studies suggest that FAT/CD36 is a potential target for O-GlcNAcylation. Conclusion/Interpretation These data demonstrate that acute modulation of HBP and protein O-GlcNAcylation in the heart stimulates fatty acid oxidation, possibly by increasing plasma membrane levels of FAT/CD36, raising the intriguing possibility that the HBP and O-GlcNAc turnover represent a novel, glucose dependent mechanism for regulating cardiac metabolism.
Collapse
|
80
|
Rajapakse AG, Ming XF, Carvas JM, Yang Z. O-linked beta-N-acetylglucosamine during hyperglycemia exerts both anti-inflammatory and pro-oxidative properties in the endothelial system. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2:172-5. [PMID: 20592773 PMCID: PMC2763244 DOI: 10.4161/oxim.2.3.8482] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 03/18/2009] [Accepted: 03/18/2009] [Indexed: 02/07/2023]
Abstract
Elevated cellular levels of protein O-linked beta-N-acetylglucosamine (O-GlcNAc) through hexosamine biosynthesis pathway (HBP) are suggested to contribute to cardiovascular adverse effects under chronic hyperglycemic condition associated with oxidative stress and inflammation. Conversely, enhancing O-GlcNAc levels have also been demonstrated being protective against myocardial ischemia/reperfusion injury. We recently demonstrated that hyperglycemia increases oxidative stress and HBP flux in endothelial cells and enhances endothelial expression of vascular adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in response to tumor necrosis factor-alpha (TNFalpha) through oxidative stress rather than HBP pathway. Here we present further complementary data showing that enhancing O-GlcNAc levels by glucosamine does not mimic hyperglycemia's effect on TNFalpha-induced endothelial VCAM-1 and ICAM-1 expression. Glucosamine however inhibits ICAM-1 (not VCAM-1) expression and induces superoxide generation in the cells. The results further suggest that increased O-GlcNAc levels do not mediate the enhancing effect of hyperglycemia on the endothelial inflammatory responses to TNFalpha. In contrast, it exerts certain anti-inflammatory effects accompanied by pro-oxidative properties. Further work should delineate the exact role of HPB pathway in different aspects of cardiovascular functions, especially those of diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Angana Gupta Rajapakse
- Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland
| | | | | | | |
Collapse
|
81
|
Issad T, Masson E, Pagesy P. O-GlcNAc modification, insulin signaling and diabetic complications. DIABETES & METABOLISM 2010; 36:423-35. [PMID: 21074472 DOI: 10.1016/j.diabet.2010.09.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 09/16/2010] [Accepted: 09/17/2010] [Indexed: 11/24/2022]
Abstract
O-GlcNAc glycosylation (O-GlcNAcylation) corresponds to the addition of N-acetylglucosamine on serine and threonine residues of cytosolic and nuclear proteins. O-GlcNAcylation is a dynamic post-translational modification, analogous to phosphorylation, that regulates the stability, the activity or the subcellular localisation of target proteins. This reversible modification depends on the availability of glucose and therefore constitutes a powerful mechanism by which cellular activities are regulated according to the nutritional environment of the cell. O-GlcNAcylation has been implicated in important human pathologies including Alzheimer disease and type-2 diabetes. Only two enzymes, OGT and O-GlcNAcase, control the O-GlcNAc level on proteins. Therefore, O-GlcNAcylations cannot organize in signaling cascades as observed for phosphorylations. O-GlcNAcylations should rather be considered as a "rheostat" that controls the intensity of the signals traveling through different pathways according to the nutritional status of the cell. Thus, OGT attenuates insulin signal by O-GlcNAcylation of proteins involved in proximal and distal steps in the PI-3 kinase signaling pathway. This negative feedback may be exacerbated when cells are chronically exposed to elevated glucose concentrations and could thereby contribute to alterations in insulin signaling observed in diabetic patients. O-GlcNAcylation also appears to contribute to the deleterious effects of hyperglycaemia on excessive glucose production by the liver and deterioration of β-cell pancreatic function, resulting in worsening of hyperglycaemia (glucotoxicity). Moreover, O-GlcNAcylations directly participate in several diabetic complications. O-GlcNAcylation of eNOS in endothelial cells have been involved in micro- and macrovascular complications. In addition, O-GlcNAcylations activate the expression of profibrotic and antifibrinolytic factors, contributing to vascular and renal dysfunctions.
Collapse
Affiliation(s)
- T Issad
- CNRS (UMR 8104), université Paris Descartes, institut Cochin, 22 rue Méchain, Paris, France.
| | | | | |
Collapse
|
82
|
Johnson PA, Luk A, Demtchouk A, Patel H, Sung HJ, Treiser MD, Gordonov S, Sheihet L, Bolikal D, Kohn J, Moghe PV. Interplay of anionic charge, poly(ethylene glycol), and iodinated tyrosine incorporation within tyrosine-derived polycarbonates: Effects on vascular smooth muscle cell adhesion, proliferation, and motility. J Biomed Mater Res A 2010; 93:505-14. [PMID: 19585568 PMCID: PMC2845728 DOI: 10.1002/jbm.a.32544] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Regulation of smooth muscle cell adhesion, proliferation, and motility on biomaterials is critical to the performance of blood-contacting implants and vascular tissue engineering scaffolds. The goal of this study was to examine the underlying substrate-smooth muscle cell response relations, using a selection of polymers representative of an expansive library of multifunctional, tyrosine-derived polycarbonates. Three chemical components within the polymer structure were selectively varied through copolymerization: (1) the content of iodinated tyrosine to achieve X-ray visibility; (2) the content of poly(ethylene glycol) (PEG) to decrease protein adsorption and cell adhesivity; and (3) the content of desaminotyrosyl-tyrosine (DT), which regulates the rate of polymer degradation. Using quartz crystal microbalance with dissipation, we quantified differential serum protein adsorption behavior because of the chemical components DT, iodinated tyrosine, and PEG: increased PEG content within the polymer structure progressively decreased protein adsorption but the simultaneous presence of both DT and iodinated tyrosine reversed the effects of PEG. The complex interplay of these components was next tested on the adhesion, proliferation, and motility behavior cultured human aortic smooth muscle cells. The incorporation of PEG into the polymer reduced cell attachment, which was reversed in the presence of iodinated tyrosine. Further, we found that as little as 10% DT content was sufficient to negate the PEG effect in polymers containing iodinated tyrosine, whereas in non-iodinated polymers, the PEG effect on cell attachment was reversed. Cross-functional analysis of motility and proliferation revealed divergent substrate chemistry related cell response regimes. For instance, within the series of polymers containing both iodinated tyrosine and 10% of DT, increasing PEG levels lowered smooth muscle cell motility without a change in the rate of cell proliferation. In contrast, for non-iodinated tyrosine and 10% of DT, increasing PEG levels increased cell proliferation significantly while reducing cell motility. Clearly, the polycarbonate polymer library offers a sensitive platform to modulate cell adhesion, proliferation, and motility responses, which, in turn, may have implications for controlling vascular remodeling in vivo. Additionally, our data suggests unique biorelevant properties following the incorporation of iodinated subunits in a polymeric biomaterial as a potential platform for X-ray visible devices.
Collapse
Affiliation(s)
- Patrick A. Johnson
- Department of Chemical and Biochemical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Arnold Luk
- Department of Biomedical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Aleksey Demtchouk
- Department of Biomedical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Hiral Patel
- Department of Biomedical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Hak-Joon Sung
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Matthew D. Treiser
- Department of Biomedical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Simon Gordonov
- Department of Biomedical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Larisa Sheihet
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Das Bolikal
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Joachim Kohn
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| | - Prabhas V. Moghe
- Department of Biomedical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
- Department of Chemical and Biochemical Engineering, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
83
|
Katare RG, Caporali A, Oikawa A, Meloni M, Emanueli C, Madeddu P. Vitamin B1 analog benfotiamine prevents diabetes-induced diastolic dysfunction and heart failure through Akt/Pim-1-mediated survival pathway. Circ Heart Fail 2010; 3:294-305. [PMID: 20107192 DOI: 10.1161/circheartfailure.109.903450] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The increasing incidence of diabetes mellitus will result in a new epidemic of heart failure unless novel treatments able to halt diabetic cardiomyopathy early in its course are introduced. This study aimed to determine whether the activity of the Akt/Pim-1 signaling pathway is altered at critical stages of diabetic cardiomyopathy and whether supplementation with vitamin B1 analog benfotiamine (BFT) helps to sustain the above prosurvival mechanism, thereby preserving cardiomyocyte viability and function. METHODS AND RESULTS Untreated streptozotocin-induced type 1 or leptin-receptor mutant type 2 diabetic mice showed diastolic dysfunction evolving to contractile impairment and cardiac dilatation and failure. BFT (70 mg/kg(-1)/d(-1)) improved diastolic and systolic function and prevented left ventricular end-diastolic pressure increase and chamber dilatation in both diabetic models. Moreover, BFT improved cardiac perfusion and reduced cardiomyocyte apoptosis and interstitial fibrosis. In hearts of untreated diabetic mice, the expression and activity of Akt/Pim-1 signaling declined along with O-N-acetylglucosamine modification of Akt, inhibition of pentose phosphate pathway, activation of oxidative stress, and accumulation of glycation end products. Furthermore, diabetes reduced pSTAT3 independently of Akt. BFT inhibited these effects of diabetes mellitus, thereby conferring cardiomyocytes with improved resistance to high glucose-induced damage. The phosphoinositide-3-kinase inhibitor LY294002 and dominant-negative Akt inhibited antiapoptotic action of BFT-induced and Pim-1 upregulation in high glucose-challenged cardiomyocytes. CONCLUSIONS These results show that BFT protects from diabetes mellitus-induced cardiac dysfunction through pleiotropic mechanisms, culminating in the activation of prosurvival signaling pathway. Thus, BFT merits attention for application in clinical practice.
Collapse
Affiliation(s)
- Rajesh G Katare
- Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, United Kingdom
| | | | | | | | | | | |
Collapse
|
84
|
Lima VV, Rigsby CS, Hardy DM, Webb RC, Tostes RC. O-GlcNAcylation: a novel post-translational mechanism to alter vascular cellular signaling in health and disease: focus on hypertension. JOURNAL OF THE AMERICAN SOCIETY OF HYPERTENSION : JASH 2009; 3:374-87. [PMID: 20409980 PMCID: PMC3022480 DOI: 10.1016/j.jash.2009.09.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 09/26/2009] [Accepted: 09/28/2009] [Indexed: 12/21/2022]
Abstract
O-Linked attachment of beta-N-acetyl-glucosamine (O-GlcNAc) on serine and threonine residues of nuclear and cytoplasmic proteins is a highly dynamic posttranslational modification that plays a key role in signal transduction pathways. Preliminary data show that O-GlcNAcylation may represent a key regulatory mechanism in the vasculature, modulating contractile and relaxant responses. Proteins with an important role in vascular function, such as endothelial nitric oxide synthase, sarcoplasmic reticulum Ca(2+)-ATPase, protein kinase C, mitogen-activated protein kinases, and proteins involved in cytoskeleton regulation and microtubule assembly are targets for O-GlcNAcylation, indicating that this posttranslational modification may play an important role in vascular reactivity. Here, we will focus on a few specific pathways that contribute to vascular function and cardiovascular disease-associated vascular dysfunction, and the implications of their modification by O-GlcNAc. New chemical tools have been developed to detect and study O-GlcNAcylation, including inhibitors of O-GlcNAc enzymes, chemoenzymatic tagging methods, and quantitative proteomics strategies; these will also be briefly addressed. An exciting challenge in the future will be to better understand the cellular dynamics of this posttranslational modification, as well as the signaling pathways and mechanisms by which O-GlcNAc is regulated on specific proteins in the vasculature in health and disease.
Collapse
Affiliation(s)
- Victor V. Lima
- Department of Physiology, Medical College of Georgia, Augusta, GA, USA
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - David M. Hardy
- Department of Physiology, Medical College of Georgia, Augusta, GA, USA
- Department of Surgery, Medical College of Georgia, Augusta, GA, USA
| | - R. Clinton Webb
- Department of Physiology, Medical College of Georgia, Augusta, GA, USA
| | - Rita C. Tostes
- Department of Physiology, Medical College of Georgia, Augusta, GA, USA
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
85
|
D'Souza DR, Salib MM, Bennett J, Mochin-Peters M, Asrani K, Goldblum SE, Renoud KJ, Shapiro P, Passaniti A. Hyperglycemia regulates RUNX2 activation and cellular wound healing through the aldose reductase polyol pathway. J Biol Chem 2009; 284:17947-55. [PMID: 19383984 PMCID: PMC2709386 DOI: 10.1074/jbc.m109.002378] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Indexed: 11/06/2022] Open
Abstract
Diabetes mellitus accelerates cardiovascular microangiopathies and atherosclerosis, which are a consequence of hyperglycemia. The aldose reductase (AR) polyol pathway contributes to these microvascular complications, but how it mediates vascular damage in response to hyperglycemia is less understood. The RUNX2 transcription factor, which is repressed in diabetic animals, promotes vascular endothelial cell (EC) migration, proliferation, and angiogenesis. Here we show that physiological levels of glucose (euglycemia) increase RUNX2 DNA binding and transcriptional activity, whereas hyperglycemia does not. However, inhibition of AR reverses hyperglycemic suppression of RUNX2. IGF-1 secretion and IGF receptor phosphorylation by autocrine IGF-1 occur equally in euglycemic or hyperglycemic conditions, suggesting that reduced RUNX2 activity in response to hyperglycemia is not because of altered IGF-1/IGF receptor activation. AR also negatively regulates RUNX2-dependent vascular remodeling in an EC wounded monolayer assay, which is reversed by specific AR inhibition in hyperglycemia. Thus, euglycemia supports RUNX2 activity and promotes normal microvascular EC migration and wound healing, which are repressed under hyperglycemic conditions through the AR polyol pathway.
Collapse
Affiliation(s)
- David R. D'Souza
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
| | - Maryann M. Salib
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
| | - Jessica Bennett
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
| | - Maria Mochin-Peters
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
| | - Kaushal Asrani
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
| | - Simeon E. Goldblum
- the Department of Medicine and
- Mucosal Biology Research Center, University of Maryland School of Medicine, and
| | - Keli J. Renoud
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
| | - Paul Shapiro
- the Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Antonino Passaniti
- From the Departments of Pathology and Biochemistry and Molecular Biology, Graduate Program in Life Sciences, Marlene and Stewart Greenebaum Cancer Center
- Mucosal Biology Research Center, University of Maryland School of Medicine, and
| |
Collapse
|
86
|
Laczy B, Hill BG, Wang K, Paterson AJ, White CR, Xing D, Chen YF, Darley-Usmar V, Oparil S, Chatham JC. Protein O-GlcNAcylation: a new signaling paradigm for the cardiovascular system. Am J Physiol Heart Circ Physiol 2009; 296:H13-28. [PMID: 19028792 PMCID: PMC2637779 DOI: 10.1152/ajpheart.01056.2008] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 11/11/2008] [Indexed: 02/07/2023]
Abstract
The posttranslational modification of serine and threonine residues of nuclear and cytoplasmic proteins by the O-linked attachment of the monosaccharide beta-N-acetylglucosamine (O-GlcNAc) is a highly dynamic and ubiquitous protein modification. Protein O-GlcNAcylation is rapidly emerging as a key regulator of critical biological processes including nuclear transport, translation and transcription, signal transduction, cytoskeletal reorganization, proteasomal degradation, and apoptosis. Increased levels of O-GlcNAc have been implicated as a pathogenic contributor to glucose toxicity and insulin resistance, which are both major hallmarks of diabetes mellitus and diabetes-related cardiovascular complications. Conversely, there is a growing body of data demonstrating that the acute activation of O-GlcNAc levels is an endogenous stress response designed to enhance cell survival. Reports on the effect of altered O-GlcNAc levels on the heart and cardiovascular system have been growing rapidly over the past few years and have implicated a role for O-GlcNAc in contributing to the adverse effects of diabetes on cardiovascular function as well as mediating the response to ischemic injury. Here, we summarize our present understanding of protein O-GlcNAcylation and its effect on the regulation of cardiovascular function. We examine the pathways regulating protein O-GlcNAcylation and discuss, in more detail, our understanding of the role of O-GlcNAc in both mediating the adverse effects of diabetes as well as its role in mediating cellular protective mechanisms in the cardiovascular system. In addition, we also explore the parallels between O-GlcNAc signaling and redox signaling, as an alternative paradigm for understanding the role of O-GlcNAcylation in regulating cell function.
Collapse
Affiliation(s)
- Boglarka Laczy
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-0007, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Issad T, Kuo M. O-GlcNAc modification of transcription factors, glucose sensing and glucotoxicity. Trends Endocrinol Metab 2008; 19:380-9. [PMID: 18929495 DOI: 10.1016/j.tem.2008.09.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 09/05/2008] [Accepted: 09/08/2008] [Indexed: 10/21/2022]
Abstract
Regulation of proteins by O-GlcNAc modification is becoming a major area of research. This reversible modification depends on glucose concentrations and, therefore, constitutes a powerful mechanism to regulate protein activities according to glucose availability. Its importance in glucose-dependent gene transcription is underlined by its role in pancreatic insulin biosynthesis (through PDX-1 and NeuroD1 O-GlcNAc modifications) and leptin synthesis in adipose tissue (through Sp1 O-GlcNAc modification). Moreover, in chronic hyperglycaemia, O-GlcNAc modifications of Sp1, p53 and NFkappaB participate in glucotoxicity, resulting in cardiovascular and renal alterations. The recent discovery by two independent groups that FoxO1 is regulated by O-GlcNAc modification provides a potential mechanism by which hyperglycaemia promotes gluconeogenesis and worsening of glucose intolerance, opening new research perspectives in the field.
Collapse
Affiliation(s)
- Tarik Issad
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France.
| | | |
Collapse
|
88
|
Ngoh GA, Jones SP. New insights into metabolic signaling and cell survival: the role of beta-O-linkage of N-acetylglucosamine. J Pharmacol Exp Ther 2008; 327:602-9. [PMID: 18768779 PMCID: PMC6545568 DOI: 10.1124/jpet.108.143263] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The involvement of glucose in fundamental metabolic pathways represents a core element of biology. Late in the 20th century, a unique glucose-derived signal was discovered, which appeared to be involved in a variety of cellular processes, including mitosis, transcription, insulin signaling, stress responses, and potentially, Alzheimer's disease, and diabetes. By definition, this glucose-fed signaling system was a post-translational modification to proteins. However, unlike classical cotranslational N-glycosylation occurring in the endoplasmic reticulum and Golgi apparatus, this process occurs elsewhere throughout the cell in a highly dynamic fashion, similar to the quintessential post-translational modification, phosphorylation. This more recently described post-translational modification, the beta-O-linkage of N-acetylglucosamine (i.e., O-GlcNAc) to nucleocytoplasmic proteins, represents an under-investigated area of biology. This signaling system operates in all of the tissues examined and seems to have persisted throughout all multicellular eukaryotes. Thus, it comes with little surprise that O-GlcNAc signaling is an integral system and viable target for biomedical investigation. This system may be a boundless source for insight into a variety of diseases and yield numerous opportunities for drug design. This Perspective will address recent insights into O-GlcNAc signaling in the cardiovascular system as a paradigm for its involvement in other biological systems.
Collapse
Affiliation(s)
- Gladys A Ngoh
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | |
Collapse
|
89
|
Soesanto YA, Luo B, Jones D, Taylor R, Gabrielsen JS, Parker G, McClain DA. Regulation of Akt signaling by O-GlcNAc in euglycemia. Am J Physiol Endocrinol Metab 2008; 295:E974-80. [PMID: 18728220 PMCID: PMC2575895 DOI: 10.1152/ajpendo.90366.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The hexosamine biosynthesis pathway (HBP) regulates the posttranslational modification of nuclear and cytoplasmic protein by O-linked N-acetylglucosamine (O-GlcNAc). Numerous studies have demonstrated that, in hyperglycemic conditions, excessive glucose flux through this pathway contributes to the development of insulin resistance. The role of the HBP in euglycemia, however, remains largely unknown. Here we investigated the effect of O-GlcNAc on hepatic Akt signaling at physiological concentrations of glucose. In HepG2 cells cultured in 5 mM glucose, removal of O-GlcNAc by adenoviral-mediated overexpression of O-GlcNAcase increased Akt activity and phosphorylation. We also observed that Akt was recognized by succinylated wheat germ agglutinin (sWGA), which specifically binds O-GlcNAc. Overexpression of O-GlcNAcase in HepG2 cells reduced the levels of Akt in sWGA precipitates. The increased Akt activity was accompanied by increased phosphorylation of Akt substrates and reduced mRNA for glucose-6-phosphatase and phosphoenolpyruvate carboxykinase (PEPCK). The increased Akt activity was not a result of activation of its upstream activator phosphoinositide 3-kinase (PI 3-kinase). Further demonstrating Akt regulation by O-GlcNAc, we found that overexpression of O-GlcNAcase in the livers of euglycemic mice also significantly increased Akt activity, resulting in increased phosphorylation of downstream targets and decreased mRNA for glucose-6-phosphatase. Together, these data suggest that O-GlcNAc regulates Akt signaling in hepatic models under euglycemic conditions.
Collapse
Affiliation(s)
- Yudi A Soesanto
- Division of Endocrinology, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Rask-Madsen C, King GL. More sugar, less blood vessels: another piece in the puzzle of increased cardiovascular risk in diabetes. Arterioscler Thromb Vasc Biol 2008; 28:608-10. [PMID: 18354093 PMCID: PMC3232058 DOI: 10.1161/atvbaha.108.162057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|