1
|
Ruzic M, Hefferan AIA, Conaway A, Hogan DA. Pseudomonas aeruginosa LasR-deficient mutants have increased methylglyoxal and hydrogen peroxide sensitivity due to low intracellular glutathione. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615034. [PMID: 39386711 PMCID: PMC11463435 DOI: 10.1101/2024.09.25.615034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The electrophile methylglyoxal (MG) is produced by microorganisms and host cells through central metabolic pathways. MG is a highly reactive electrophile, so it must be rapidly detoxified to prevent damaging modifications to macromolecules. Pseudomonas aeruginosa, a pathogen of concern due to its ability develop multidrug resistance, causes many types of infections that have been associated with elevated MG levels, including cystic fibrosis (CF). P. aeruginosa isolates commonly have mutations that lead to LasR loss-of-function (LasR-) and we found that lasR mutations confer sensitivity to MG in multiple strain backgrounds. LasR- strains have increased activity of the CbrAB two-component system which represses Crc regulation of metabolism. Here, we show that higher CbrAB activity and low Crc activity renders cells sensitive to MG. We found that P. aeruginosa LasR- strains are more sensitive to MG and have lower intracellular reduced glutathione (GSH) compared to their LasR+ comparators. Consistent with published reports, mutants lacking gloA3, which encodes a MG-glyoxalase, and mutants lacking GSH biosynthesis enzymes (gshA or gshB) were sensitive to MG. Exogenous GSH rescued MG sensitivity in LasR- strains and gshA or gshB mutants, but not in a gloA3 mutant strain. We propose that low GSH levels in LasR- strains contribute to increased sensitivity to MG and H2O2.
Collapse
Affiliation(s)
- Marina Ruzic
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | - Amy Conaway
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Deborah A Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
2
|
Yang C, Zhu Q, Chen Y, Ji K, Li S, Wu Q, Pan Q, Li J. Review of the Protective Mechanism of Curcumin on Cardiovascular Disease. Drug Des Devel Ther 2024; 18:165-192. [PMID: 38312990 PMCID: PMC10838105 DOI: 10.2147/dddt.s445555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death worldwide and has been the focus of research in the medical community. Curcumin is a polyphenolic compound extracted from the root of turmeric. Curcumin has been shown to have a variety of pharmacological properties over the past decades. Curcumin can significantly protect cardiomyocyte injury after ischemia and hypoxia, inhibit myocardial hypertrophy and fibrosis, improve ventricular remodeling, reduce drug-induced myocardial injury, improve diabetic cardiomyopathy(DCM), alleviate vascular endothelial dysfunction, inhibit foam cell formation, and reduce vascular smooth muscle cells(VSMCs) proliferation. Clinical studies have shown that curcumin has a protective effect on blood vessels. Toxicological studies have shown that curcumin is safe. But high doses of curcumin also have some side effects, such as liver damage and defects in embryonic heart development. This article reviews the mechanism of curcumin intervention on CVDs in recent years, in order to provide reference for the development of new drugs in the future.
Collapse
Affiliation(s)
- Chunkun Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Yanbo Chen
- Department of Arrhythmia, Weifang People's Hospital, Weifang, Shandong, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Qian Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
3
|
Berdowska I, Matusiewicz M, Fecka I. Methylglyoxal in Cardiometabolic Disorders: Routes Leading to Pathology Counterbalanced by Treatment Strategies. Molecules 2023; 28:7742. [PMID: 38067472 PMCID: PMC10708463 DOI: 10.3390/molecules28237742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Methylglyoxal (MGO) is the major compound belonging to reactive carbonyl species (RCS) responsible for the generation of advanced glycation end products (AGEs). Its upregulation, followed by deleterious effects at the cellular and systemic levels, is associated with metabolic disturbances (hyperglycemia/hyperinsulinemia/insulin resistance/hyperlipidemia/inflammatory processes/carbonyl stress/oxidative stress/hypoxia). Therefore, it is implicated in a variety of disorders, including metabolic syndrome, diabetes mellitus, and cardiovascular diseases. In this review, an interplay between pathways leading to MGO generation and scavenging is addressed in regard to this system's impairment in pathology. The issues associated with mechanistic MGO involvement in pathological processes, as well as the discussion on its possible causative role in cardiometabolic diseases, are enclosed. Finally, the main strategies aimed at MGO and its AGEs downregulation with respect to cardiometabolic disorders treatment are addressed. Potential glycation inhibitors and MGO scavengers are discussed, as well as the mechanisms of their action.
Collapse
Affiliation(s)
- Izabela Berdowska
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | | | - Izabela Fecka
- Department of Pharmacognosy and Herbal Medicines, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
4
|
Albrecht M, Sticht C, Wagner T, Hettler SA, De La Torre C, Qiu J, Gretz N, Albrecht T, Yard B, Sleeman JP, Garvalov BK. The crosstalk between glomerular endothelial cells and podocytes controls their responses to metabolic stimuli in diabetic nephropathy. Sci Rep 2023; 13:17985. [PMID: 37863933 PMCID: PMC10589299 DOI: 10.1038/s41598-023-45139-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
In diabetic nephropathy (DN), glomerular endothelial cells (GECs) and podocytes undergo pathological alterations, which are influenced by metabolic changes characteristic of diabetes, including hyperglycaemia (HG) and elevated methylglyoxal (MGO) levels. However, it remains insufficiently understood what effects these metabolic factors have on GEC and podocytes and to what extent the interactions between the two cell types can modulate these effects. To address these questions, we established a co-culture system in which GECs and podocytes were grown together in close proximity, and assessed transcriptional changes in each cell type after exposure to HG and MGO. We found that HG and MGO had distinct effects on gene expression and that the effect of each treatment was markedly different between GECs and podocytes. HG treatment led to upregulation of "immediate early response" genes, particularly those of the EGR family, as well as genes involved in inflammatory responses (in GECs) or DNA replication/cell cycle (in podocytes). Interestingly, both HG and MGO led to downregulation of genes related to extracellular matrix organisation in podocytes. Crucially, the transcriptional responses of GECs and podocytes were dependent on their interaction with each other, as many of the prominently regulated genes in co-culture of the two cell types were not significantly changed when monocultures of the cells were exposed to the same stimuli. Finally, the changes in the expression of selected genes were validated in BTBR ob/ob mice, an established model of DN. This work highlights the molecular alterations in GECs and podocytes in response to the key diabetic metabolic triggers HG and MGO, as well as the central role of GEC-podocyte crosstalk in governing these responses.
Collapse
Affiliation(s)
- Michael Albrecht
- European Center for Angioscience (ECAS), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany
| | - Carsten Sticht
- Center of Medical Research, Bioinformatics and Statistics, Medical Faculty Mannheim of the University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- NGS Core Facility, Medical Faculty Mannheim of the University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Tabea Wagner
- European Center for Angioscience (ECAS), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany
| | - Steffen A Hettler
- Department of Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology and Pneumology, Fifth Department of Medicine, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Carolina De La Torre
- Center of Medical Research, Bioinformatics and Statistics, Medical Faculty Mannheim of the University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- NGS Core Facility, Medical Faculty Mannheim of the University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Jiedong Qiu
- Department of Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology and Pneumology, Fifth Department of Medicine, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Center of Medical Research, Bioinformatics and Statistics, Medical Faculty Mannheim of the University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Thomas Albrecht
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Benito Yard
- Department of Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology and Pneumology, Fifth Department of Medicine, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Jonathan P Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany.
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany.
- Institute of Biological and Chemical Systems - Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology Campus North, Building 319, Hermann-Von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
| | - Boyan K Garvalov
- European Center for Angioscience (ECAS), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany.
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim of the University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
5
|
Alyahyawi AR, Khan MY, Alouffi S, Maarfi F, Akasha R, Khan S, Rafi Z, Alharazi T, Shahab U, Ahmad S. Identification of Glycoxidative Lesion in Isolated Low-Density Lipoproteins from Diabetes Mellitus Subjects. Life (Basel) 2023; 13:1986. [PMID: 37895368 PMCID: PMC10608319 DOI: 10.3390/life13101986] [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: 08/16/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 10/29/2023] Open
Abstract
Methylglyoxal (MG) is a precursor for advanced glycation end-products (AGEs), which have a significant role in diabetes. The present study is designed to probe the immunological response of native and glycated low-density lipoprotein (LDL) in experimental animals. The second part of this study is to probe glycoxidative lesion detection in low-density lipoproteins (LDL) in diabetes subjects with varying disease duration. The neo-epitopes attributed to glycation-induced glycoxidative lesion of LDL in DM patients' plasma were, analyzed by binding of native and MG-modified LDL immunized animal sera antibodies using an immunochemical assay. The plasma purified human LDL glycation with MG, which instigated modification in LDL. Further, the NewZealand-White rabbits were infused with unmodified natural LDL (N-LDL) and MG-glycatedLDL to probe its immunogenicity. The glycoxidative lesion detection in LDL of DM with disease duration (D.D.) of 5-15 years and D.D. > 15 years was found to be significantly higher as compared to normal healthy subjects (NHS) LDL. The findings support the notion that prolonged duration of diabetes can cause structural alteration in LDL protein molecules, rendering them highly immunogenic in nature. The presence of LDL lesions specific to MG-associated glycoxidation would further help in assessing the progression of diabetes mellitus.
Collapse
Affiliation(s)
- Amjad R. Alyahyawi
- Department of Diagnostic Radiology, College of Applied Medical Science, University of Hail, Ha’il 2440, Saudi Arabia;
- Centre for Nuclear and Radiation Physics, Department of Physics, University of Surrey, Guildford GU2 7XH, UK
| | - Mohd Yasir Khan
- Department of Biotechnology, SALS, Uttaranchal University, Dehradun 248011, India;
| | - Sultan Alouffi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha’il 2440, Saudi Arabia; (S.A.); (R.A.); (T.A.)
| | - Farah Maarfi
- Department of Biotechnology, SALS, Uttaranchal University, Dehradun 248011, India;
| | - Rihab Akasha
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha’il 2440, Saudi Arabia; (S.A.); (R.A.); (T.A.)
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, Hail University, Ha’il 2440, Saudi Arabia;
| | - Zeeshan Rafi
- Department of Bioengineering, Integral University, Lucknow 226026, India;
| | - Talal Alharazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha’il 2440, Saudi Arabia; (S.A.); (R.A.); (T.A.)
| | - Uzma Shahab
- Department of Biochemistry, King George Medical University, Lucknow 226026, India;
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha’il 2440, Saudi Arabia; (S.A.); (R.A.); (T.A.)
| |
Collapse
|
6
|
Wang W, Ye J, Guo Z, Ma Y, Yang Q, Zhong W, Du S, Bai J. A novel glycoprotein from earthworm extract PvE-3: Insights of their characteristics for promoting diabetic wound healing and attenuating methylglyoxal-induced cell damage. Int J Biol Macromol 2023; 239:124267. [PMID: 37003377 DOI: 10.1016/j.ijbiomac.2023.124267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/18/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Diabetic chronic wound is a worldwide medical burden related to overdosed methylglyoxal (MGO) synthesis, which is the major precursor of glycation of proteins and DNA and is related to the dysfunction of dermal cells thus leading to chronic refractory wounds. Previous studies proved that earthworm extract accelerates diabetic wound healing and possesses cell proliferation and antioxidative effects. However, the effects of earthworm extract on MGO-damaged fibroblasts, the inner mechanisms of MGO-induced cell damage and the functional components in earthworm extract are still poorly understood. Firstly, we evaluated the bioactivities of the earthworm extract PvE-3 on the diabetic wound model and the diabetic related cell damage model. Then the mechanisms were investigated through transcriptomics, flow cytometry and fluorescence probe. The results revealed that PvE-3 promoted diabetic wound healing and protected fibroblast function in cell-damaged conditions. Meanwhile, the high-throughput screening implied the inner mechanisms of diabetic wound healing and PvE-3 cytoprotection effect were involved in the muscle cell function, the cell cycle regulation and the mitochondrial transmembrane potential depolarization. The functional glycoprotein isolated from PvE-3 possessed EGF-like domain which had a strong binding affinity with EGFR. The findings provided references to explore the potential treatments of diabetic wound healing.
Collapse
Affiliation(s)
- Wenjie Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinhong Ye
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zishuo Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yunnan Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qilin Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wanling Zhong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shouying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Jie Bai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
7
|
De Masi R, Orlando S. GANAB and N-Glycans Substrates Are Relevant in Human Physiology, Polycystic Pathology and Multiple Sclerosis: A Review. Int J Mol Sci 2022; 23:7373. [PMID: 35806376 PMCID: PMC9266668 DOI: 10.3390/ijms23137373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Glycans are one of the four fundamental macromolecular components of living matter, and they are highly regulated in the cell. Their functions are metabolic, structural and modulatory. In particular, ER resident N-glycans participate with the Glc3Man9GlcNAc2 highly conserved sequence, in protein folding process, where the physiological balance between glycosylation/deglycosylation on the innermost glucose residue takes place, according GANAB/UGGT concentration ratio. However, under abnormal conditions, the cell adapts to the glucose availability by adopting an aerobic or anaerobic regimen of glycolysis, or to external stimuli through internal or external recognition patterns, so it responds to pathogenic noxa with unfolded protein response (UPR). UPR can affect Multiple Sclerosis (MS) and several neurological and metabolic diseases via the BiP stress sensor, resulting in ATF6, PERK and IRE1 activation. Furthermore, the abnormal GANAB expression has been observed in MS, systemic lupus erythematous, male germinal epithelium and predisposed highly replicating cells of the kidney tubules and bile ducts. The latter is the case of Polycystic Liver Disease (PCLD) and Polycystic Kidney Disease (PCKD), where genetically induced GANAB loss affects polycystin-1 (PC1) and polycystin-2 (PC2), resulting in altered protein quality control and cyst formation phenomenon. Our topics resume the role of glycans in cell physiology, highlighting the N-glycans one, as a substrate of GANAB, which is an emerging key molecule in MS and other human pathologies.
Collapse
Affiliation(s)
- Roberto De Masi
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy;
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| |
Collapse
|
8
|
ApoA-I Nanoparticles as Curcumin Carriers for Cerebral Endothelial Cells: Improved Cytoprotective Effects against Methylglyoxal. Pharmaceuticals (Basel) 2022; 15:ph15030347. [PMID: 35337146 PMCID: PMC8952315 DOI: 10.3390/ph15030347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 11/29/2022] Open
Abstract
Methylglyoxal (MGO) is a highly reactive metabolite of glucose present at elevated levels in diabetic patients. Its cytotoxicity is associated with endothelial dysfunction, which plays a role in cardiovascular and cerebrovascular complications. Although curcumin has many therapeutic benefits, these are limited due to its low bioavailability. We aimed to improve the bioavailability of curcumin and evaluate a potential synergistic effect of curcumin and reconstituted high-density lipoprotein (rHDL) nanoparticles (Cur-rHDLs) on MGO-induced cytotoxicity and oxidative stress in murine cerebrovascular endothelial cells (bEnd.3). Cur-rHDL nanoparticles (14.02 ± 0.95 nm) prepared by ultracentrifugation and containing curcumin were quantified by LC–MS/MS. The synergistic effect of cur-rHDL nanoparticles was tested on bEnd.3 cytotoxicity, reactive oxygen species (ROS) production, chromatin condensation, endoplasmic reticulum (ER) stress, and endothelial barrier integrity by impedancemetry. The uptake of curcumin, alone or associated with HDLs, was also assessed by mass spectrometry. Pretreatment with Cur-rHDLs followed by incubation with MGO showed a protective effect on MGO-induced cytotoxicity and chromatin condensation, as well as a strong protective effect on ROS production, endothelial cell barrier integrity, and ER stress. These results suggest that Cur-rHDLs could be used as a potential therapeutic agent to limit MGO-induced dysfunction in cerebrovascular endothelial cells by enhancing the bioavailability and protective effects of curcumin.
Collapse
|
9
|
Monnier VM, Sell DR, Gao X, Genuth SM, Lachin JM, Bebu I. Plasma advanced glycation end products and the subsequent risk of microvascular complications in type 1 diabetes in the DCCT/EDIC. BMJ Open Diabetes Res Care 2022; 10:10/1/e002667. [PMID: 35058313 PMCID: PMC8783825 DOI: 10.1136/bmjdrc-2021-002667] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/01/2021] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION To assess impact of glycemic control on plasma protein-bound advanced glycation end products (pAGEs) and their association with subsequent microvascular disease. RESEARCH DESIGN AND METHODS Eleven pAGEs were measured by liquid chromatography-mass spectrometry in banked plasma from 466 participants in the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) study at three time points (TPs): DCCT year 4 (TP1) and year 8 (TP2) and EDIC year 5/6 (TP3). Correlation coefficients assessed cross-sectional associations, and Cox proportional hazards models assessed associations with subsequent risk of microvascular complications through EDIC year 24. RESULTS Glucose-derived glycation products fructose-lysine (FL), glucosepane (GSPN) and carboxymethyl-lysine (CML) decreased with intensive glycemic control at both TP1 and TP2 (p<0.0001) but were similar at TP3, and correlated with hemoglobin A1c (HbA1c). At TP1, the markers were associated with the subsequent risk of several microvascular outcomes. These associations did not remain significant after adjustment for HbA1c, except methionine sulfoxide (MetSOX), which remained associated with diabetic kidney disease. In unadjusted models using all 3 TPs, glucose-derived pAGEs were associated with subsequent risk of proliferative diabetic retinopathy (PDR, p<0.003), clinically significant macular edema (CSME, p<0.015) and confirmed clinical neuropathy (CCN, p<0.018, except CML, not significant (NS)). Adjusted for age, sex, body mass index, diabetes duration and mean updated HbA1c, the associations remained significant for PDR (FL: p<0.002, GSPN: p≤0.02, CML: p<0.003, pentosidine: p<0.02), CMSE (CML: p<0.03), albuminuria (FL: p<0.02, CML: p<0.03) and CCN (FL: p<0.005, GSPN : p<0.003). CONCLUSIONS pAGEs at TP1 are not superior to HbA1c for risk prediction, but glucose-derived pAGEs at three TPs and MetSOX remain robustly associated with progression of microvascular complications in type 1 diabetes even after adjustment for HbA1c and other factors.
Collapse
Affiliation(s)
- Vincent M Monnier
- Pathology and Biochemistry, Case Western Reserve University Department of Pathology, Cleveland, Ohio, USA
| | - David R Sell
- Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiaoyu Gao
- The Biostatistics Center, The George Washington University, Rockville, Maryland, USA
| | - Saul M Genuth
- Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - John M Lachin
- The Biostatistics Center, The George Washington University, Rockville, Maryland, USA
| | - Ionut Bebu
- The Biostatistics Center, The George Washington University, Rockville, Maryland, USA
| |
Collapse
|
10
|
Hernandez-Castillo C, Shuck SC. Diet and Obesity-Induced Methylglyoxal Production and Links to Metabolic Disease. Chem Res Toxicol 2021; 34:2424-2440. [PMID: 34851609 DOI: 10.1021/acs.chemrestox.1c00221] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The obesity rate in the United States is 42.4% and has become a national epidemic. Obesity is a complex condition that is influenced by socioeconomic status, ethnicity, genetics, age, and diet. Increased consumption of a Western diet, one that is high in processed foods, red meat, and sugar content, is associated with elevated obesity rates. Factors that increase obesity risk, such as socioeconomic status, also increase consumption of a Western diet because of a limited access to healthier options and greater affordability of processed foods. Obesity is a public health threat because it increases the risk of several pathologies, including atherosclerosis, diabetes, and cancer. The molecular mechanisms linking obesity to disease onset and progression are not well understood, but a proposed mechanism is physiological changes caused by altered lipid peroxidation, glycolysis, and protein metabolism. These metabolic pathways give rise to reactive molecules such as the abundant electrophile methylglyoxal (MG), which covalently modifies nucleic acids and proteins. MG-adducts are associated with obesity-linked pathologies and may have potential for biomonitoring to determine the risk of disease onset and progression. MG-adducts may also play a role in disease progression because they are mutagenic and directly impact protein stability and function. In this review, we discuss how obesity drives metabolic alterations, how these alterations lead to MG production, the association of MG-adducts with disease, and the potential impact of MG-adducts on cellular function.
Collapse
Affiliation(s)
- Carlos Hernandez-Castillo
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010, United States
| | - Sarah C Shuck
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010, United States
| |
Collapse
|
11
|
Tang SS, Xing SY, Zhang XJ, Ren XQ, Hong H, Long Y. Neuroprotective effects of novel compound Tozan on cognition, neurogenesis and apoptosis in diabetes. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1471. [PMID: 34734023 PMCID: PMC8506716 DOI: 10.21037/atm-21-4439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/10/2021] [Indexed: 11/06/2022]
Abstract
Background Cognitive impairment is a serious complication of diabetes that manifests as an impairment of spatial memory and learning ability. Its pathogenesis is unclear, and effective therapeutic drugs are very limited. Our group designed and synthesized a novel compound named 3-p-tolyl-9H-xanthen-9-one (Tozan). In this study, we sought to investigate the effects and mechanism of Tozan on diabetic cognitive impairment. Methods Methylglyoxal (MG)-induced SH-SY5Y cells and streptozotocin (STZ)-induced type 1 diabetic mice were treated with Tozan. Methyl thiazolul tetrazolium (MTT) and lactate dehydrogenase (LDH) were used to test cytotoxicity. Morris water maze (MWM) and Y-maze tests were used to evaluate cognitive function. Immunofluorescence and western blot analyses were used to evaluate neurogenesis, apoptosis, and signal transduction pathway-related proteins. In addition, Lentivirus (LV)-estrogen receptor beta (ERβ)-ribonucleic acid interference (RNAi) was used to knockdown the ERβ gene in SH-SY5Y cells. Results We found that Tozan ameliorated MG-induced cytotoxicity in SH-SY5Y cells, improved cognitive dysfunction in STZ-induced type 1 diabetic mice, increased neurogenesis, and prevented apoptotic responses in vitro and in vivo. Importantly, Tozan (2, 4, and 8 mg/kg) mediated phosphatidylinositol-3-kinase and protein kinase B cAMP-response element binding protein (PI3K/Akt-CREB) signaling by activating membrane ERβ, and a high dose of Tozan (8 mg/kg) mediated CREB signaling by activating nuclear ERβ in the hippocampus. Notably, Tozan did not have an anti-apoptosis and regeneration protective role in ERβ gene knockdown cells. Conclusions Our study demonstrates Tozan’s contributions to and role in cognition, neurogenesis, and apoptosis in diabetes, and lays an experimental foundation for the development of new anti-diabetic cognitive impairment drugs.
Collapse
Affiliation(s)
- Su-Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Shu-Yun Xing
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Xue-Jiao Zhang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Xiao-Qian Ren
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Yan Long
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
12
|
Saeed M, Kausar MA, Singh R, Siddiqui AJ, Akhter A. The Role of Glyoxalase in Glycation and Carbonyl Stress Induced Metabolic Disorders. Curr Protein Pept Sci 2021; 21:846-859. [PMID: 32368974 DOI: 10.2174/1389203721666200505101734] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/09/2019] [Accepted: 12/08/2019] [Indexed: 12/14/2022]
Abstract
Glycation refers to the covalent binding of sugar molecules to macromolecules, such as DNA, proteins, and lipids in a non-enzymatic reaction, resulting in the formation of irreversibly bound products known as advanced glycation end products (AGEs). AGEs are synthesized in high amounts both in pathological conditions, such as diabetes and under physiological conditions resulting in aging. The body's anti-glycation defense mechanisms play a critical role in removing glycated products. However, if this defense system fails, AGEs start accumulating, which results in pathological conditions. Studies have been shown that increased accumulation of AGEs acts as key mediators in multiple diseases, such as diabetes, obesity, arthritis, cancer, atherosclerosis, decreased skin elasticity, male erectile dysfunction, pulmonary fibrosis, aging, and Alzheimer's disease. Furthermore, glycation of nucleotides, proteins, and phospholipids by α-oxoaldehyde metabolites, such as glyoxal (GO) and methylglyoxal (MGO), causes potential damage to the genome, proteome, and lipidome. Glyoxalase-1 (GLO-1) acts as a part of the anti-glycation defense system by carrying out detoxification of GO and MGO. It has been demonstrated that GLO-1 protects dicarbonyl modifications of the proteome and lipidome, thereby impeding the cell signaling and affecting age-related diseases. Its relationship with detoxification and anti-glycation defense is well established. Glycation of proteins by MGO and GO results in protein misfolding, thereby affecting their structure and function. These findings provide evidence for the rationale that the functional modulation of the GLO pathway could be used as a potential therapeutic target. In the present review, we summarized the newly emerged literature on the GLO pathway, including enzymes regulating the process. In addition, we described small bioactive molecules with the potential to modulate the GLO pathway, thereby providing a basis for the development of new treatment strategies against age-related complications.
Collapse
Affiliation(s)
- Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Hail, Saudi Arabia
| | - Rajeev Singh
- Department of Environmental Studies, Sataywati College, Delhi University, Delhi, India
| | - Arif J Siddiqui
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Asma Akhter
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 226026, India
| |
Collapse
|
13
|
Dicarbonyl derived post-translational modifications: chemistry bridging biology and aging-related disease. Essays Biochem 2020; 64:97-110. [PMID: 31939602 DOI: 10.1042/ebc20190057] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/17/2023]
Abstract
In living systems, nucleophilic amino acid residues are prone to non-enzymatic post-translational modification by electrophiles. α-Dicarbonyl compounds are a special type of electrophiles that can react irreversibly with lysine, arginine, and cysteine residues via complex mechanisms to form post-translational modifications known as advanced glycation end-products (AGEs). Glyoxal, methylglyoxal, and 3-deoxyglucosone are the major endogenous dicarbonyls, with methylglyoxal being the most well-studied. There are several routes that lead to the formation of dicarbonyl compounds, most originating from glucose and glucose metabolism, such as the non-enzymatic decomposition of glycolytic intermediates and fructosyl amines. Although dicarbonyls are removed continuously mainly via the glyoxalase system, several conditions lead to an increase in dicarbonyl concentration and thereby AGE formation. AGEs have been implicated in diabetes and aging-related diseases, and for this reason the elucidation of their structure as well as protein targets is of great interest. Though the dicarbonyls and reactive protein side chains are of relatively simple nature, the structures of the adducts as well as their mechanism of formation are not that trivial. Furthermore, detection of sites of modification can be demanding and current best practices rely on either direct mass spectrometry or various methods of enrichment based on antibodies or click chemistry followed by mass spectrometry. Future research into the structure of these adducts and protein targets of dicarbonyl compounds may improve the understanding of how the mechanisms of diabetes and aging-related physiological damage occur.
Collapse
|
14
|
Pipino C, Shah H, Prudente S, Di Pietro N, Zeng L, Park K, Trischitta V, Pennathur S, Pandolfi A, Doria A. Association of the 1q25 Diabetes-Specific Coronary Heart Disease Locus With Alterations of the γ-Glutamyl Cycle and Increased Methylglyoxal Levels in Endothelial Cells. Diabetes 2020; 69:2206-2216. [PMID: 32651240 PMCID: PMC7506838 DOI: 10.2337/db20-0475] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022]
Abstract
A chromosome 1q25 variant (rs10911021) has been associated with coronary heart disease (CHD) in type 2 diabetes. In human umbilical vein endothelial cells (HUVECs), the risk allele "C" is associated with lower expression of the adjacent gene GLUL encoding glutamine synthase, converting glutamic acid to glutamine. To further investigate the mechanisms through which this locus affects CHD risk, we measured 35 intracellular metabolites involved in glutamic acid metabolism and the γ-glutamyl cycle in 62 HUVEC strains carrying different rs10911021 genotypes. Eight metabolites were positively associated with the risk allele (17-58% increase/allele copy, P = 0.046-0.002), including five γ-glutamyl amino acids, β-citryl-glutamate, N-acetyl-aspartyl-glutamate, and ophthalmate-a marker of γ-glutamyl cycle malfunction. Consistent with these findings, the risk allele was also associated with decreased glutathione-to-glutamate ratio (-9%, P = 0.012), decreased S-lactoylglutathione (-41%, P = 0.019), and reduced detoxification of the atherogenic compound methylglyoxal (+54%, P = 0.008). GLUL downregulation by shRNA caused a 40% increase in the methylglyoxal level, which was completely prevented by glutamine supplementation. In summary, we have identified intracellular metabolic traits associated with the 1q25 risk allele in HUVECs, including impairments of the γ-glutamyl cycle and methylglyoxal detoxification. Glutamine supplementation abolishes the latter abnormality, suggesting that such treatment may prevent CHD in 1q25 risk allele carriers.
Collapse
Affiliation(s)
- Caterina Pipino
- Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology - CAST (ex CeSI-MeT), University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Hetal Shah
- Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Sabrina Prudente
- Research Unit of Metabolic and Cardiovascular Diseases, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Natalia Di Pietro
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology - CAST (ex CeSI-MeT), University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Lixia Zeng
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Kyoungmin Park
- Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Subramanian Pennathur
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology - CAST (ex CeSI-MeT), University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Alessandro Doria
- Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Schalkwijk CG, Stehouwer CDA. Methylglyoxal, a Highly Reactive Dicarbonyl Compound, in Diabetes, Its Vascular Complications, and Other Age-Related Diseases. Physiol Rev 2020; 100:407-461. [DOI: 10.1152/physrev.00001.2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The formation and accumulation of methylglyoxal (MGO), a highly reactive dicarbonyl compound, has been implicated in the pathogenesis of type 2 diabetes, vascular complications of diabetes, and several other age-related chronic inflammatory diseases such as cardiovascular disease, cancer, and disorders of the central nervous system. MGO is mainly formed as a byproduct of glycolysis and, under physiological circumstances, detoxified by the glyoxalase system. MGO is the major precursor of nonenzymatic glycation of proteins and DNA, subsequently leading to the formation of advanced glycation end products (AGEs). MGO and MGO-derived AGEs can impact on organs and tissues affecting their functions and structure. In this review we summarize the formation of MGO, the detoxification of MGO by the glyoxalase system, and the biochemical pathways through which MGO is linked to the development of diabetes, vascular complications of diabetes, and other age-related diseases. Although interventions to treat MGO-associated complications are not yet available in the clinical setting, several strategies to lower MGO have been developed over the years. We will summarize several new directions to target MGO stress including glyoxalase inducers and MGO scavengers. Targeting MGO burden may provide new therapeutic applications to mitigate diseases in which MGO plays a crucial role.
Collapse
Affiliation(s)
- C. G. Schalkwijk
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - C. D. A. Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
16
|
Psoralea corylifolia L. Seed Extract Attenuates Methylglyoxal-Induced Insulin Resistance by Inhibition of Advanced Glycation End Product Formation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4310319. [PMID: 31976027 PMCID: PMC6954480 DOI: 10.1155/2019/4310319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/31/2019] [Accepted: 12/05/2019] [Indexed: 01/11/2023]
Abstract
Accumulation of advanced glycation end products (AGEs) in the body has been implicated in the pathogenesis of metabolic conditions, such as diabetes mellitus. Methylglyoxal (MGO), a major precursor of AGEs, has been reported to induce insulin resistance in both in vitro and in vivo studies. Psoralea corylifolia seeds (PCS) have been used as a traditional medicine for several diseases, but their potential application in treating insulin resistance has not yet been evaluated. This study is aimed at investigating whether PCS extract could attenuate insulin resistance induced by MGO. Male C57BL/6N mice (6 weeks old) were administered 1% MGO in their drinking water for 18 weeks, and the PCS extract (200 or 500 mg/kg) was orally administered daily from the first day of the MGO administration. We observed that both 200 and 500 mg/kg PCS extract treatment significantly improved glucose tolerance and insulin sensitivity and markedly restored p-Akt and p-IRS1/2 expression in the livers of the MGO-administered mice. Additionally, the PCS extract significantly increased the phosphorylation of Akt and IRS-1/2 and glucose uptake in MGO-treated HepG2 cells. Further studies showed that the PCS extract inhibited MGO-induced AGE formation in the HepG2 cells and in the sera of MGO-administered mice. PCS extract also increased the expression of glyoxalase 1 (GLO1) in the liver tissue of MGO-administered mice. The PCS extract significantly decreased the phosphorylation of ERK, p38, and NF-κB and suppressed the mRNA expression of proinflammatory molecules including TNF-α and IL-1β and iNOS in MGO-administered mice. Additionally, we demonstrated that the PCS extract attenuated oxidative stress, as evidenced by the reduced ROS production in the MGO-treated cells and the enhanced expression of antioxidant enzymes in the liver of MGO-administered mice. Thus, PCS extract ameliorated the MGO-induced insulin resistance in HepG2 cells and in mice by reducing oxidative stress via the inhibition of AGE formation. These findings suggest the potential of PCS extract as a candidate for the prevention and treatment of insulin resistance.
Collapse
|
17
|
Advanced glycation endproducts produced by in vitro glycation of type I collagen modulate the functional and secretory behavior of dorsal root ganglion cells cultivated in two-dimensional system. Exp Cell Res 2019; 382:111475. [PMID: 31255600 DOI: 10.1016/j.yexcr.2019.06.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/15/2019] [Accepted: 06/19/2019] [Indexed: 12/24/2022]
Abstract
Advanced glycation end-products (AGEs) are proteins/lipids that are glycated upon sugar exposure and are often increased during inflammatory diseases such as osteoarthritis and neurodegenerative disorders. Here, we developed an extracellular matrix (ECM) using glycated type I collagen (ECM-GC), which produced similar levels of AGEs to those detected in the sera of arthritic mice. In order to determine whether AGEs were sufficient to stimulate sensory neurons, dorsal root ganglia (DRGs) cells were cultured on ECM-GC or ECM-NC-coated plates. ECM-GC or ECM-NC were favorable for DRG cells expansion. However, ECM-GC cultivated neurons displayed thinner F-actin filaments, rounded morphology, and reduced neuron interconnection compared to ECM-NC. In addition, ECM-GC did not affect RAGE expression levels in the neurons, although induced rapid p38, MAPK and ERK activation. Finally, ECM-GC stimulated the secretion of nitrite and TNF-α by DRG cells. Taken together, our in vitro glycated ECM model suitably mimics the in vivo microenvironment of inflammatory disorders and provides new insights into the role of ECM impairment as a nociceptive stimulus.
Collapse
|
18
|
Wetzels S, Vanmierlo T, Scheijen JLJM, van Horssen J, Amor S, Somers V, Schalkwijk CG, Hendriks JJA, Wouters K. Methylglyoxal-Derived Advanced Glycation Endproducts Accumulate in Multiple Sclerosis Lesions. Front Immunol 2019; 10:855. [PMID: 31068938 PMCID: PMC6491451 DOI: 10.3389/fimmu.2019.00855] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/02/2019] [Indexed: 11/18/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease in which innate and adaptive immune cells infiltrate the central nervous system (CNS) and damage the myelin sheaths surrounding the axons. Upon activation, infiltrated macrophages, CNS-resident microglia, and astrocytes switch their metabolism toward glycolysis, resulting in the formation of α-dicarbonyls, such as methylglyoxal (MGO) and glyoxal (GO). These potent glycating agents lead to the formation of advanced glycation endproducts (AGEs) after reaction with amino acids. We hypothesize that AGE levels are increased in MS lesions due to the inflammatory activation of macrophages and astrocytes. First, we measured tissue levels of AGEs in brain samples of MS patients and controls. Analysis of MS patient and non-demented control (NDC) specimens showed a significant increase in protein-bound Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1), the major AGE, compared to white matter of NDCs (107 ± 11 vs. 154 ± 21, p < 0.05). In addition, immunohistochemistry revealed that MGO-derived AGEs were specifically present in astrocytes, whereas the receptor for AGEs, RAGE, was detected on microglia/macrophages. Moreover, in cerebrospinal fluid from MS patients, α-dicarbonyls and free AGEs correlated with their respective levels in the plasma, whereas this was not observed for protein-bound AGEs. Taken together, our data show that MG-H1 is produced by astrocytes. This suggests that AGEs secreted by astrocytes have paracrine effects on RAGE-positive macrophages/microglia and thereby contribute to the pathology of MS.
Collapse
Affiliation(s)
- Suzan Wetzels
- Cardiovascular Research Institute Maastricht, Department of Internal Medicine, Maastricht University, Maastricht, Netherlands.,Biomedical Research Institute, Department of Immunology and Biochemistry, Hasselt University, Hasselt, Belgium
| | - Tim Vanmierlo
- Biomedical Research Institute, Department of Immunology and Biochemistry, Hasselt University, Hasselt, Belgium.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Jean L J M Scheijen
- Cardiovascular Research Institute Maastricht, Department of Internal Medicine, Maastricht University, Maastricht, Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
| | - Veerle Somers
- Biomedical Research Institute, Department of Immunology and Biochemistry, Hasselt University, Hasselt, Belgium
| | - Casper G Schalkwijk
- Cardiovascular Research Institute Maastricht, Department of Internal Medicine, Maastricht University, Maastricht, Netherlands
| | - Jerome J A Hendriks
- Biomedical Research Institute, Department of Immunology and Biochemistry, Hasselt University, Hasselt, Belgium
| | - Kristiaan Wouters
- Cardiovascular Research Institute Maastricht, Department of Internal Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
19
|
Ravichandran G, Lakshmanan DK, Raju K, Elangovan A, Nambirajan G, Devanesan AA, Thilagar S. Food advanced glycation end products as potential endocrine disruptors: An emerging threat to contemporary and future generation. ENVIRONMENT INTERNATIONAL 2019; 123:486-500. [PMID: 30622074 DOI: 10.1016/j.envint.2018.12.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 06/09/2023]
Abstract
Mankind exposure to chemicals in the past century has increased dramatically throughout environment. There is no question that chemicals interfere with the physiology of biological system. Abundance of chemicals is documented to be detrimental to human and wildlife. The mammalian endocrine system is comprised of many interacting tissues mediate themselves through hormones that are essential for metabolism, growth and development. Humans secrete over fifty different hormones to orchestrate major physiological functions however; these vital functions can be intervened by huge number of internal and external chemical stressors that are identified as endocrine disruptors. Advanced glycation end products (AGEs), familiarly known as Maillard products, formed through non-enzymatic glycation whose production is augmented on aging as well as environmental stressors. Processed foods have become very popular today due to their taste, convenience, and inexpensiveness. Manufacture of these day-to-day foods involves extreme temperatures on processing results in the formation of AGEs could independently promote oxidative stress, aging, diabetes, cancer, degenerative diseases, more fascinatingly hormonal disruption is the subject of interest of this review. Based on some substantial observations documented till time, we discuss the emergence of dietary AGEs as potential endocrine disruptors by emphasizing their occurrence, mechanisms and participation in endocrine interruption. Both economically and in terms of human life, AGEs may represent an enormous cost for the future society. Therefore, by explicating their novel role in endocrine diseases, the review strives to make an impact on AGEs and their exposure among public as well as scientific communities.
Collapse
Affiliation(s)
- Guna Ravichandran
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Dinesh Kumar Lakshmanan
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Karthik Raju
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Abbirami Elangovan
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Gayathri Nambirajan
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Arul Ananth Devanesan
- Department of Food Quality and Safety, Gilat Research Center, Agricultural Research Organization, M.P. Negev 85280, Israel
| | - Sivasudha Thilagar
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India.
| |
Collapse
|
20
|
Shamsaldeen YA, Alsugoor MH, Lione LA, Benham CD. Dysfunction in nitric oxide synthesis in streptozotocin treated rat aorta and role of methylglyoxal. Eur J Pharmacol 2019; 842:321-328. [DOI: 10.1016/j.ejphar.2018.10.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022]
|
21
|
Schumacher D, Morgenstern J, Oguchi Y, Volk N, Kopf S, Groener JB, Nawroth PP, Fleming T, Freichel M. Compensatory mechanisms for methylglyoxal detoxification in experimental & clinical diabetes. Mol Metab 2018; 18:143-152. [PMID: 30287091 PMCID: PMC6308908 DOI: 10.1016/j.molmet.2018.09.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES The deficit of Glyoxalase I (Glo1) and the subsequent increase in methylglyoxal (MG) has been reported to be one the five mechanisms by which hyperglycemia causes diabetic late complications. Aldo-keto reductases (AKR) have been shown to metabolize MG; however, the relative contribution of this superfamily to the detoxification of MG in vivo, particularly within the diabetic state, remains unknown. METHODS CRISPR/Cas9-mediated genome editing was used to generate a Glo1 knock-out (Glo1-/-) mouse line. Streptozotocin was then applied to investigate metabolic changes under hyperglycemic conditions. RESULTS Glo1-/- mice were viable and showed no elevated MG or MG-H1 levels under hyperglycemic conditions. It was subsequently found that the enzymatic efficiency of various oxidoreductases in the liver and kidney towards MG were increased in the Glo1-/- mice. The functional relevance of this was supported by the altered distribution of alternative detoxification products. Furthermore, it was shown that MG-dependent AKR activity is a potentially clinical relevant pathway in human patients suffering from diabetes. CONCLUSIONS These data suggest that in the absence of GLO1, AKR can effectively compensate to prevent the accumulation of MG. The combination of metabolic, enzymatic, and genetic factors, therefore, may provide a better means of identifying patients who are at risk for the development of late complications caused by elevated levels of MG.
Collapse
Affiliation(s)
- Dagmar Schumacher
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Yoko Oguchi
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Nadine Volk
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Stefan Kopf
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jan Benedikt Groener
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter Paul Nawroth
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Joint Division Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg Center for Molecular Biology (ZMBH), Heidelberg, Germany; University Hospital Heidelberg University, Heidelberg, Germany; Germany Institute for Diabetes, Neuherberg, Germany; Cancer IDC Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Neuherberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marc Freichel
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
22
|
Wetzels S, Wouters K, Miyata T, Scheijen JLJM, Hendriks JJA, Schalkwijk CG, Vanmierlo T. Advanced Glycation Endproducts Are Increased in the Animal Model of Multiple Sclerosis but Cannot Be Reduced by Pyridoxamine Treatment or Glyoxalase 1 Overexpression. Int J Mol Sci 2018; 19:ijms19051311. [PMID: 29702605 PMCID: PMC5983766 DOI: 10.3390/ijms19051311] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS). The immune response in MS patients leads to the infiltration of immune cells in the CNS and their subsequent activation. Immune cell activation induces a switch towards glycolysis. During glycolysis, the dicarbonyl product methylglyoxal (MGO) is produced. MGO is a glycating agent that can rapidly form advanced glycation endproducts (AGEs). In turn, AGEs are able to induce inflammatory responses. The glyoxalase system is the endogenous defense system of the body to reduce the burden of MGO thereby reducing AGE formation. This system consists of glyoxalase-1 and glyoxalase-2 which are able to detoxify MGO to D-lactate. We investigated whether AGE levels are induced in experimental autoimmune encephalitis (EAE), an inflammatory animal model of MS. Twenty seven days post EAE induction, MGO and AGE (Nε-(carboxymethyl)lysine (CML), Nε-(carboxyethyl)lysine (CEL), 5-hydro-5-methylimidazolone (MG-H1)) levels were significantly increased in the spinal cord of mice subjected to EAE. Yet, pyridoxamine treatment and glyoxalase-1 overexpression were unable to counteract AGE production during EAE and did not influence the clinical course of EAE. In conclusion, AGEs levels increase during EAE in the spinal cord, but AGE-modifying treatments do not inhibit EAE-induced AGE production and do not affect disease progression.
Collapse
Affiliation(s)
- Suzan Wetzels
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Martelarenlaan 42 3500 Hasselt, Belgium.
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Kristiaan Wouters
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Toshio Miyata
- Division of Molecular Medicine and Therapy, Tohoku University Graduate School of Medicine, 980-8577 Sendai, Japan.
| | - Jean L J M Scheijen
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Jerome J A Hendriks
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Martelarenlaan 42 3500 Hasselt, Belgium.
| | - Casper G Schalkwijk
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Tim Vanmierlo
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Martelarenlaan 42 3500 Hasselt, Belgium.
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
23
|
Chen XM, Zhang WQ, Tian Y, Wang LF, Chen CC, Qiu CM. Liraglutide suppresses non-esterified free fatty acids and soluble vascular cell adhesion molecule-1 compared with metformin in patients with recent-onset type 2 diabetes. Cardiovasc Diabetol 2018; 17:53. [PMID: 29636047 PMCID: PMC5891985 DOI: 10.1186/s12933-018-0701-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/05/2018] [Indexed: 01/28/2023] Open
Abstract
Background It has been suggested that liraglutide could have an impact on glucose and lipid metabolism disorder and adhesion molecule activation, which may play important roles in the vascular damage of diabetes. In this study, we examined the effects of liraglutide versus metformin on non-esterified free fatty acids, beta-cell insulin secretion, and adhesion molecule levels in patients with recent-onset type 2 diabetes mellitus. Methods In this study, 60 patients newly diagnosed with type 2 diabetes mellitus (mean age 33.97 ± 5.67 years) were randomly assigned to receive once-daily subcutaneous liraglutide or oral metformin. Before the study and after the 8-week treatment period, a 75 g oral glucose tolerance test was performed. Plasma glucose, lipids and lipoprotein, plasma insulin, glycaemic and insulin responses, non-esterified free fatty acids (NEFA), and soluble vascular cell adhesion molecule-1 (sVCAM-1) levels were evaluated. Results After 8 weeks, 120 min of NEFA (155 ± 125 vs 99 ± 73 µmol/L, P = 0.026) and the levels of sVCAM-1 (465 ± 136 vs 382 ± 131 ng/ml, P = 0.013) significantly decreased, while the early phase insulin secretion index (24.94 [7.78, 38.89] vs. 31.13 [17.67, 59.09], P = 0.031), fasting plasma insulin (104 [51, 123] vs 113 [54, 171] mIU/L, P = 0.015), 60 min plasma insulin (326 [165, 441] vs 471 [334, 717] mIU/L, P = 0.005), 120 min plasma insulin (401 [193, 560] vs 500 [367, 960] mIU/L, P = 0.047), and insulin area under the curve (AUCins) (648 [321, 742] vs 738 [451, 1118] mIU/L, P = 0.005) remarkably increased for patients in the liraglutide treatment group. The levels of sVCAM-1 dramatically decreased after 8 weeks of liraglutide treatment (503 ± 182 vs 382 ± 131 ng/ml, P = 0.046) compared to that of the metformin treatment group. At the same time, the differences before and after liraglutide treatment in 120 min of NEFA (− 32 [− 96, − 5] vs 5 [− 35, 38] µmol/L, P = 0.033) and AUCins (738 [451, 1118] vs 594 [357, 1216] mIU/L, P = 0.014) were remarkably enhanced compared to that of the metformin therapy. Nevertheless, there were no significant differences in fasting NEFA after liraglutide or metformin treatment. The reduction of 120 min NEFA (ΔNEFA) was positively correlated with the decrease of sVCAM-1 (ΔsVCAM-1) after 8 weeks of liraglutide treatment (r = 0.523, P = 0.003). Conclusions Our results demonstrate that liraglutide administration is more effective than metformin in reducing 120 min NEFA and suppressing sVCAM-1 levels for recent-onset type 2 diabetes mellitus. We suggest that this outcome may be because liraglutide is associated with potentiating insulin secretion capacity, inhibiting vascular inflammatory cytokines, and antagonizing atherosclerosis. Electronic supplementary material The online version of this article (10.1186/s12933-018-0701-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao-Min Chen
- Department of Endocrinology and Metabolism, Zhongshan Hospital Xiamen University, 201-209 Hubin South Road, Xiamen, 361004, People's Republic of China.
| | - Wen-Qiang Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital Xiamen University, 201-209 Hubin South Road, Xiamen, 361004, People's Republic of China
| | - Yuan Tian
- Department of Endocrinology and Metabolism, Zhongshan Hospital Xiamen University, 201-209 Hubin South Road, Xiamen, 361004, People's Republic of China
| | - Li-Fen Wang
- Guangzhou Medicine University Second Affiliated Hospital, 250-296 Changgang East Road, Guangzhou, 510260, People's Republic of China
| | - Chan-Chan Chen
- Department of Endocrinology and Metabolism, Zhongshan Hospital Xiamen University, 201-209 Hubin South Road, Xiamen, 361004, People's Republic of China
| | - Chuan-Mei Qiu
- Department of Endocrinology and Metabolism, Zhongshan Hospital Xiamen University, 201-209 Hubin South Road, Xiamen, 361004, People's Republic of China
| |
Collapse
|
24
|
Methylglyoxal: A Relevant Marker of Disease Activity in Patients with Rheumatoid Arthritis. DISEASE MARKERS 2018; 2018:8735926. [PMID: 29606988 PMCID: PMC5828101 DOI: 10.1155/2018/8735926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/01/2018] [Indexed: 12/29/2022]
Abstract
Background The contribution of methylglyoxal (MGO) and soluble receptor for advanced glycation end products (sRAGE) in the presence of rheumatoid arthritis (RA) is still unknown. We investigated whether serum MGO and sRAGE were related to the presence of disease activity in RA. Methods 80 patients with RA and 30 control subjects were included in a cross-sectional study. The severity of RA was assessed using the disease activity score for 28 joints (DAS28). Serum MGO and sRAGE were measured by ELISA. Results Serum MGO levels were significantly higher in patients with RA versus control subjects (P < 0.001) and were increased in RA patients with higher disease activity versus RA patients with moderate disease activity (P = 0.019). Serum sRAGE concentrations were significantly decreased in RA patients with higher disease activity versus RA patients with moderate disease activity and versus control subjects (P = 0.004; P = 0.002, resp.). A multiple logistic regression analysis demonstrated that MGO was independently associated with the presence of activity disease in RA (OR = 1.17, 95% CI: 1.02–1.31, P = 0.01). Conclusion Serum MGO and sRAGE levels are inversely related to the activity of RA, and MGO is independently associated with a higher disease activity of RA.
Collapse
|
25
|
Hanssen NMJ, Scheijen JLJM, Jorsal A, Parving HH, Tarnow L, Rossing P, Stehouwer CDA, Schalkwijk CG. Higher Plasma Methylglyoxal Levels Are Associated With Incident Cardiovascular Disease in Individuals With Type 1 Diabetes: A 12-Year Follow-up Study. Diabetes 2017; 66:2278-2283. [PMID: 28588100 DOI: 10.2337/db16-1578] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/21/2017] [Indexed: 11/13/2022]
Abstract
Methylglyoxal (MGO), a major precursor for advanced glycation end products, is increased in diabetes. In diabetic rodents, inhibition of MGO prevents cardiovascular disease (CVD). Whether plasma MGO levels are associated with incident CVD in people with type 1 diabetes is unknown. We included 159 individuals with persistent normoalbuminuria and 162 individuals with diabetic nephropathy (DN) from the outpatient clinic at Steno Diabetes Center. We measured MGO at baseline and recorded fatal and nonfatal CVD over a median follow-up of 12.3 years (interquartile range 7.6-12.5 years). Data were analyzed by Cox regression, with adjustment for sex, age, HbA1c, DN, diabetes duration, smoking, systolic blood pressure, antihypertensive medication, and BMI. During follow-up, 73 individuals suffered at least one CVD event (36 fatal and 53 nonfatal). Higher MGO levels were associated with total, fatal, and nonfatal incident CVD (hazard ratios [HRs] 1.47 [95% CI 1.13-1.91], 1.42 [1.01-1.99], and 1.46 [1.08-1.98], respectively). We observed a similar trend for total mortality (HR 1.24 [0.99-1.56]). This study shows for the first time in our knowledge that plasma MGO levels are associated with cardiovascular events in individuals with type 1 diabetes. MGO may explain, at least in part, the increased risk for CVD in type 1 diabetes.
Collapse
Affiliation(s)
- Nordin M J Hanssen
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Jean L J M Scheijen
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Anders Jorsal
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Hans-Henrik Parving
- Department of Medical Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health Science, Aarhus University, Aarhus, Denmark
- Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Lise Tarnow
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Faculty of Health Science, Aarhus University, Aarhus, Denmark
- Nordsjællands Hospital, Hillerød, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Faculty of Health Science, Aarhus University, Aarhus, Denmark
- Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| |
Collapse
|
26
|
Are targeted therapies for diabetic cardiomyopathy on the horizon? Clin Sci (Lond) 2017; 131:897-915. [PMID: 28473471 DOI: 10.1042/cs20160491] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/02/2017] [Accepted: 02/03/2017] [Indexed: 12/12/2022]
Abstract
Diabetes increases the risk of heart failure approximately 2.5-fold, independent of coronary artery disease and other comorbidities. This process, termed diabetic cardiomyopathy, is characterized by initial impairment of left ventricular (LV) relaxation followed by LV contractile dysfunction. Post-mortem examination reveals that human diastolic dysfunction is closely associated with LV damage, including cardiomyocyte hypertrophy, apoptosis and fibrosis, with impaired coronary microvascular perfusion. The pathophysiological mechanisms underpinning the characteristic features of diabetic cardiomyopathy remain poorly understood, although multiple factors including altered lipid metabolism, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, inflammation, as well as epigenetic changes, are implicated. Despite a recent rise in research interrogating these mechanisms and an increased understanding of the clinical importance of diabetic cardiomyopathy, there remains a lack of specific treatment strategies. How the chronic metabolic disturbances observed in diabetes lead to structural and functional changes remains a pertinent question, and it is hoped that recent advances, particularly in the area of epigenetics, among others, may provide some answers. This review hence explores the temporal onset of the pathological features of diabetic cardiomyopathy, and their relative contribution to the resultant disease phenotype, as well as both current and potential therapeutic options. The emergence of glucose-optimizing agents, namely glucagon-like peptide-1 (GLP-1) agonists and sodium/glucose co-transporter (SGLT)2 inhibitors that confer benefits on cardiovascular outcomes, together with novel experimental approaches, highlight a new and exciting era in diabetes research, which is likely to result in major clinical impact.
Collapse
|
27
|
Dicarbonyls and Advanced Glycation End-Products in the Development of Diabetic Complications and Targets for Intervention. Int J Mol Sci 2017; 18:ijms18050984. [PMID: 28475116 PMCID: PMC5454897 DOI: 10.3390/ijms18050984] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/11/2017] [Accepted: 05/02/2017] [Indexed: 01/17/2023] Open
Abstract
Advanced glycation end-products (AGEs) are non-enzymatic protein and amino acid adducts as well as DNA adducts which form from dicarbonyls and glucose. AGE formation is enhanced in diabetes and is associated with the development of diabetic complications. In the current review, we discuss mechanisms that lead to enhanced AGE levels in the context of diabetes and diabetic complications. The methylglyoxal-detoxifying glyoxalase system as well as alternative pathways of AGE detoxification are summarized. Therapeutic approaches to interfere with different pathways of AGE formation are presented.
Collapse
|
28
|
Matafome P, Rodrigues T, Sena C, Seiça R. Methylglyoxal in Metabolic Disorders: Facts, Myths, and Promises. Med Res Rev 2017; 37:368-403. [PMID: 27636890 DOI: 10.1002/med.21410] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/07/2016] [Accepted: 08/12/2016] [Indexed: 08/26/2024]
Abstract
Glucose and fructose metabolism originates the highly reactive byproduct methylglyoxal (MG), which is a strong precursor of advanced glycation end products (AGE). The MG has been implicated in classical diabetic complications such as retinopathy, nephropathy, and neuropathy, but has also been recently associated with cardiovascular diseases and central nervous system disorders such as cerebrovascular diseases and dementia. Recent studies even suggested its involvement in insulin resistance and beta-cell dysfunction, contributing to the early development of type 2 diabetes and creating a vicious circle between glycation and hyperglycemia. Despite several drugs and natural compounds have been identified in the last years in order to scavenge MG and inhibit AGE formation, we are still far from having an effective strategy to prevent MG-induced mechanisms. This review summarizes the endogenous and exogenous sources of MG, also addressing the current controversy about the importance of exogenous MG sources. The mechanisms by which MG changes cell behavior and its involvement in type 2 diabetes development and complications and the pathophysiological implication are also summarized. Particular emphasis will be given to pathophysiological relevance of studies using higher MG doses, which may have produced biased results. Finally, we also overview the current knowledge about detoxification strategies, including modulation of endogenous enzymatic systems and exogenous compounds able to inhibit MG effects on biological systems.
Collapse
Affiliation(s)
- Paulo Matafome
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
- Department of Complementary Sciences, Coimbra Health School (ESTeSC), Instituto Politécnico de Coimbra, 3045-601, Coimbra, Portugal
| | - Tiago Rodrigues
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Cristina Sena
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Raquel Seiça
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| |
Collapse
|
29
|
Wetzels S, Wouters K, Schalkwijk CG, Vanmierlo T, Hendriks JJA. Methylglyoxal-Derived Advanced Glycation Endproducts in Multiple Sclerosis. Int J Mol Sci 2017; 18:ijms18020421. [PMID: 28212304 PMCID: PMC5343955 DOI: 10.3390/ijms18020421] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/23/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS). The activation of inflammatory cells is crucial for the development of MS and is shown to induce intracellular glycolytic metabolism in pro-inflammatory microglia and macrophages, as well as CNS-resident astrocytes. Advanced glycation endproducts (AGEs) are stable endproducts formed by a reaction of the dicarbonyl compounds methylglyoxal (MGO) and glyoxal (GO) with amino acids in proteins, during glycolysis. This suggests that, in MS, MGO-derived AGEs are formed in glycolysis-driven cells. MGO and MGO-derived AGEs can further activate inflammatory cells by binding to the receptor for advanced glycation endproducts (RAGE). Recent studies have revealed that AGEs are increased in the plasma and brain of MS patients. Therefore, AGEs might contribute to the inflammatory status in MS. Moreover, the main detoxification system of dicarbonyl compounds, the glyoxalase system, seems to be affected in MS patients, which may contribute to high MGO-derived AGE levels. Altogether, evidence is emerging for a contributing role of AGEs in the pathology of MS. In this review, we provide an overview of the current knowledge on the involvement of AGEs in MS.
Collapse
Affiliation(s)
- Suzan Wetzels
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 Maastricht, The Netherlands.
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium.
| | - Kristiaan Wouters
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 Maastricht, The Netherlands.
| | - Casper G Schalkwijk
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 Maastricht, The Netherlands.
| | - Tim Vanmierlo
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium.
| | - Jerome J A Hendriks
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium.
| |
Collapse
|
30
|
Methylglyoxal-Glyoxalase 1 Balance: The Root of Vascular Damage. Int J Mol Sci 2017; 18:ijms18010188. [PMID: 28106778 PMCID: PMC5297820 DOI: 10.3390/ijms18010188] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/20/2022] Open
Abstract
The highly reactive dicarbonyl methylglyoxal (MGO) is mainly formed as byproduct of glycolysis. Therefore, high blood glucose levels determine increased MGO accumulation. Nonetheless, MGO levels are also increased as consequence of the ineffective action of its main detoxification pathway, the glyoxalase system, of which glyoxalase 1 (Glo1) is the rate-limiting enzyme. Indeed, a physiological decrease of Glo1 transcription and activity occurs not only in chronic hyperglycaemia but also with ageing, during which MGO accumulation occurs. MGO and its advanced glycated end products (AGEs) are associated with age-related diseases including diabetes, vascular dysfunction and neurodegeneration. Endothelial dysfunction is the first step in the initiation, progression and clinical outcome of vascular complications, such as retinopathy, nephropathy, impaired wound healing and macroangiopathy. Because of these considerations, studies have been centered on understanding the molecular basis of endothelial dysfunction in diabetes, unveiling a central role of MGO-Glo1 imbalance in the onset of vascular complications. This review focuses on the current understanding of MGO accumulation and Glo1 activity in diabetes, and their contribution on the impairment of endothelial function leading to diabetes-associated vascular damage.
Collapse
|
31
|
Adams JN, Martelle SE, Raffield LM, Freedman BI, Langefeld CD, Hsu FC, Maldjian JA, Williamson JD, Hugenschmidt CE, Carr JJ, Cox AJ, Bowden DW. Analysis of advanced glycation end products in the DHS Mind Study. J Diabetes Complications 2016; 30:262-8. [PMID: 26739237 PMCID: PMC4761276 DOI: 10.1016/j.jdiacomp.2015.11.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/16/2015] [Accepted: 11/29/2015] [Indexed: 12/11/2022]
Abstract
AIMS Human studies of links between advanced glycation end-products (AGEs) and disease phenotypes are less common than studies of animal and cell models. Here, we examined the association of total AGEs with diabetes risk factors in a predominately type 2 diabetes (T2D) affected cohort. METHODS AGEs were measured using an enzyme linked immunosorbant assay in 816 individuals from the DHS Mind Study (n=709 T2D affected), and association analyses were completed. RESULTS Total AGEs were associated with estimated glomerular filtration rate (p=0.0054; β=-0.1291) and coronary artery calcification (p=0.0352; β=1.1489) in the entire cohort. No significant associations were observed when individuals with T2D were analyzed separately. In individuals without T2D, increased circulating AGEs were associated with increased BMI (p=0.02, β=0.138), low density lipoproteins (p=0.046, β=17.07) and triglycerides (p=0.0004, β=0.125), and decreased carotid artery calcification (p=0.0004, β=-1.2632) and estimated glomerular filtration rate (p=0.0018, β=-0.1405). Strong trends were also observed for an association between AGEs and poorer cognitive performance on the digit symbol substitution test (p=0.046, β=-6.64) and decreased grey matter volume (p=0.037, β=-14.87). CONCLUSIONS AGEs may play an important role in a number of phenotypes and diseases, although not necessarily in interindividual variation in people with T2D. Further evaluation of specific AGE molecules may shed more light on these relationships.
Collapse
Affiliation(s)
- Jeremy N Adams
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - Susan E Martelle
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - Laura M Raffield
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - Barry I Freedman
- Department of Internal Medicine, Nephrology, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - Carl D Langefeld
- Department of Biostatistical Sciences, Wake Forest University Health Sciences, Winston Salem, North Carolina
| | - Fang-Chi Hsu
- Department of Biostatistical Sciences, Wake Forest University Health Sciences, Winston Salem, North Carolina
| | - Joseph A Maldjian
- Radiologic Sciences and Advanced NeuroScience Imaging (ANSIR) Laboratory, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jeff D Williamson
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - Christina E Hugenschmidt
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - J Jeffery Carr
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston - Salem, NC, USA
| | - Amanda J Cox
- Molecular Basis of Disease, Griffith University, Southport, QLD, Australia
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston - Salem, NC, USA.
| |
Collapse
|
32
|
Adams JN, Raffield LM, Martelle SE, Freedman BI, Langefeld CD, Carr JJ, Cox AJ, Bowden DW. Genetic analysis of advanced glycation end products in the DHS MIND study. Gene 2016; 584:173-9. [PMID: 26915486 DOI: 10.1016/j.gene.2016.02.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/21/2016] [Accepted: 02/19/2016] [Indexed: 11/24/2022]
Abstract
Advanced glycation end-products (AGEs) are a diverse group of molecules produced by the non-enzymatic addition of glucose to proteins, lipids, and nucleic acids. AGE levels have been associated with hyperglycemia and diabetic complications, especially in animal models, but less clearly in human studies. We measured total serum AGEs using an enzyme linked immunosorbant assay (ELISA) in 506 subjects from 246 families in the Diabetes Heart Study (DHS)/DHS MIND Study (n=399 type 2 diabetes (T2D)-affected). Single nucleotide polymorphisms (SNPs) in several candidate genes, including known AGE receptors, were tested for their influence on circulating AGE levels. The genetic analysis was expanded to include an exploratory genome-wide association study (GWAS) and exome chip analysis of AGEs (≈440,000 SNPs). AGEs were found to be highly heritable (h(2)=0.628, p=8.96 × 10(-10)). While no SNPs from candidate genes were significantly associated after Bonferroni correction, rs1035798 in the gene AGER was the most significantly associated (p=0.007). Additionally, rs7198427, in MT1A, showed a nominally significant p-value (p=0.0099). No SNPs from the GWAS or exome studies were identified after correction for multiple comparisons; however, rs17054480 in the PALLD2 gene on chromosome 4 showed the strongest association (p=7.77 × 10(-7)). Five SNPs at two loci (ISCA2/NPC2 and FBXO33) had p-values of less than 2.0 × 10(-5) and three additional SNPs (rs716326 in MACROD2, and rs6795197 and rs6765857 in ZBTB38) showed a nominal association with p-values of less than 1.0 × 10(-5).These findings provide a foundation for further investigation into the genetic component of circulating AGEs.
Collapse
Affiliation(s)
- Jeremy N Adams
- Program in Molecular Genetics and Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Laura M Raffield
- Program in Molecular Genetics and Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Susan E Martelle
- Integrative Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Barry I Freedman
- Department of Internal Medicine - Nephrology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Carl D Langefeld
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - J Jeffrey Carr
- Department of Radiologic Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Amanda J Cox
- Molecular Basis of Disease, Griffith University, Southport, QLD, Australia
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
33
|
Mapanga RF, Essop MF. Damaging effects of hyperglycemia on cardiovascular function: spotlight on glucose metabolic pathways. Am J Physiol Heart Circ Physiol 2016; 310:H153-73. [DOI: 10.1152/ajpheart.00206.2015] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/27/2015] [Indexed: 12/13/2022]
Abstract
The incidence of cardiovascular complications associated with hyperglycemia is a growing global health problem. This review discusses the link between hyperglycemia and cardiovascular diseases onset, focusing on the role of recently emerging downstream mediators, namely, oxidative stress and glucose metabolic pathway perturbations. The role of hyperglycemia-mediated activation of nonoxidative glucose pathways (NOGPs) [i.e., the polyol pathway, hexosamine biosynthetic pathway, advanced glycation end products (AGEs), and protein kinase C] in this process is extensively reviewed. The proposal is made that there is a unique interplay between NOGPs and a downstream convergence of detrimental effects that especially affect cardiac endothelial cells, thereby contributing to contractile dysfunction. In this process the AGE pathway emerges as a crucial mediator of hyperglycemia-mediated detrimental effects. In addition, a vicious metabolic cycle is established whereby hyperglycemia-induced NOGPs further fuel their own activation by generating even more oxidative stress, thereby exacerbating damaging effects on cardiac function. Thus NOGP inhibition, and particularly that of the AGE pathway, emerges as a novel therapeutic intervention for the treatment of cardiovascular complications such as acute myocardial infarction in the presence hyperglycemia.
Collapse
Affiliation(s)
- Rudo F. Mapanga
- Cardio-Metabolic Research Group, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - M. Faadiel Essop
- Cardio-Metabolic Research Group, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
34
|
Sun YP, Gu JF, Tan XB, Wang CF, Jia XB, Feng L, Liu JP. Curcumin inhibits advanced glycation end product-induced oxidative stress and inflammatory responses in endothelial cell damage via trapping methylglyoxal. Mol Med Rep 2015; 13:1475-86. [PMID: 26718010 PMCID: PMC4732849 DOI: 10.3892/mmr.2015.4725] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 09/03/2015] [Indexed: 11/05/2022] Open
Abstract
Methylglyoxal (MGO)-induced carbonyl stress and pro-inflammatory responses have been suggested to contribute to endothelial dysfunction. Curcumin (Cur), a polyphenolic compound from Curcuma longa L., may protect endothelial cells against carbonyl stress-induced damage by trapping dicarbonyl compounds such as MGO. However, Cur-MGO adducts have not been studied in depth to date and it remains to be known whether Cur-MGO adducts are able to attenuate endothelial damage by trapping MGO. In the present study, 1,2-diaminobenzene was reacted with MGO to ensure the reliability of the reaction system. Cur was demonstrated to trap MGO at a 1:1 ratio to form adducts 1, 2 and 3 within 720 min. The structures of these adducts were identified by high-performance liquid chromatography/electrospray ionization tandem mass spectrometry. The kinetic curves of Cur (10(-7), 10(-6) and 10(-5) M) were measured from 0-168 h by fluorescent intensity. Cur significantly inhibited the formation of advanced glycation end products (AGEs). The differences in oxidative damage and the levels of pro-inflammatory cytokines following MGO + HSA or Cur-MGO treatment were investigated in human umbilical vein endothelial cells (HUVECs). Exposure of HUVECs to the Cur-MGO reaction adducts significantly reduced the intracellular ROS levels and improved cell viability compared with MGO alone. Furthermore, there was a significant reduction in the expression levels of transforming growth factor-β1 and intercellular adhesion molecule(-1) following treatment with Cur-MGO adducts compared with MGO alone. These results provide further evidence that the trapping of MGO by Cur inhibits the formation of AGEs. The current study indicates that the protective effect of Cur on carbonyl stress and pro-inflammatory responses in endothelial damage occurs via the trapping of MGO.
Collapse
Affiliation(s)
- Yan Ping Sun
- Department of Pharmacognosy, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Jun Fei Gu
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Xiao Bin Tan
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Chun Fei Wang
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Xiao Bin Jia
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Liang Feng
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Ji Ping Liu
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, P.R. China
| |
Collapse
|
35
|
Lee SI, Patel M, Jones CM, Narendran P. Cardiovascular disease and type 1 diabetes: prevalence, prediction and management in an ageing population. Ther Adv Chronic Dis 2015; 6:347-74. [PMID: 26568811 DOI: 10.1177/2040622315598502] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) is a major cause of mortality in type 1 diabetes mellitus (T1D). However, evidence of its risks and management is often extrapolated from studies in type 2 diabetic (T2D) patients or the general population. This approach is unsatisfactory given that the underlying pathology, demographics and natural history of the disease differ between T1D and T2D. Furthermore, with a rising life expectancy, a greater number of T1D patients are exposed to the cardiovascular (CV) risk factors associated with an ageing population. The aim of this review is to examine the existing literature around CVD in T1D. We pay particular attention to CVD prevalence, how well we manage risk, potential biomarkers, and whether the studies included the older aged patients (defined as aged over 65). We also discuss approaches to the management of CV risk in the older aged. The available data suggest a significant CVD burden in patients with T1D and poor management of CV risk factors. This is underpinned by a poor evidence base for therapeutic management of CV risk specifically for patients with T1D, and in the most relevant population - the older aged patients. We would suggest that important areas remain to be addressed, particularly exploring the risks and benefits of therapeutic approaches to CVD management in the older aged.
Collapse
Affiliation(s)
- Siang Ing Lee
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Mitesh Patel
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Christopher M Jones
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Parth Narendran
- Institute of Biomedical Research, The Medical School, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
36
|
The role of methylglyoxal and the glyoxalase system in diabetes and other age-related diseases. Clin Sci (Lond) 2015; 128:839-61. [PMID: 25818485 DOI: 10.1042/cs20140683] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The formation and accumulation of advanced glycation endproducts (AGEs) are related to diabetes and other age-related diseases. Methylglyoxal (MGO), a highly reactive dicarbonyl compound, is the major precursor in the formation of AGEs. MGO is mainly formed as a byproduct of glycolysis. Under physiological circumstances, MGO is detoxified by the glyoxalase system into D-lactate, with glyoxalase I (GLO1) as the key enzyme in the anti-glycation defence. New insights indicate that increased levels of MGO and the major MGO-derived AGE, methylglyoxal-derived hydroimidazolone 1 (MG-H1), and dysfunctioning of the glyoxalase system are linked to several age-related health problems, such as diabetes, cardiovascular disease, cancer and disorders of the central nervous system. The present review summarizes the mechanisms through which MGO is formed, its detoxification by the glyoxalase system and its effect on biochemical pathways in relation to the development of age-related diseases. Although several scavengers of MGO have been developed over the years, therapies to treat MGO-associated complications are not yet available for application in clinical practice. Small bioactive inducers of GLO1 can potentially form the basis for new treatment strategies for age-related disorders in which MGO plays a pivotal role.
Collapse
|
37
|
Hanssen NMJ, Beulens JWJ, van Dieren S, Scheijen JLJM, van der A DL, Spijkerman AMW, van der Schouw YT, Stehouwer CDA, Schalkwijk CG. Plasma advanced glycation end products are associated with incident cardiovascular events in individuals with type 2 diabetes: a case-cohort study with a median follow-up of 10 years (EPIC-NL). Diabetes 2015; 64:257-65. [PMID: 24848072 DOI: 10.2337/db13-1864] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Experimental data suggest a role for advanced glycation end products (AGEs) in cardiovascular disease (CVD), particularly in type 2 diabetes (T2DM). However, epidemiological evidence of an association between high plasma AGEs and increased cardiovascular risk remains inconclusive. Therefore, in a case-cohort study comprising 134 cardiovascular case subjects and a random subcohort of 218 individuals (including 65 cardiovascular case subjects), all with T2DM and nested in the European Prospective Investigation into Cancer and Nutrition in the Netherlands (EPIC-NL) study, plasma levels of protein-bound Nε-(carboxymethyl)lysine, Nε-(carboxyethyl)lysine, and pentosidine were measured with liquid chromatography. AGEs were loge-transformed, combined in a z-score, and the association with incident cardiovascular events was analyzed with Cox proportional hazard regression, adapted for case-cohort design (Prentice method). After multivariable adjustment (sex, age, cohort status, diabetes duration, total cholesterol to HDL-cholesterol ratio, smoking, systolic blood pressure, BMI, blood pressure-, cholesterol- and glucose-lowering treatment, prior cardiovascular events, and triglycerides), higher plasma AGE z-scores were associated with higher risk of incident cardiovascular events in individuals without prior cardiovascular events (hazard ratio 1.31 [95% CI: 1.06-1.61]). A similar trend was observed in individuals with prior cardiovascular events (1.37 [0.63-2.98]). In conclusion, high plasma AGEs were associated with incident cardiovascular events in individuals with T2DM. These results underline the potential importance of AGEs in development of CVD.
Collapse
Affiliation(s)
- Nordin M J Hanssen
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Joline W J Beulens
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Susan van Dieren
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jean L J M Scheijen
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Daphne L van der A
- The National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | | | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Coen D A Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
38
|
Figarola JL, Singhal J, Rahbar S, Awasthi S, Singhal SS. LR-90 prevents methylglyoxal-induced oxidative stress and apoptosis in human endothelial cells. Apoptosis 2014; 19:776-88. [PMID: 24615331 DOI: 10.1007/s10495-014-0974-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Methylglyoxal (MGO) is a highly reactive dicarbonyl compound known to induce cellular injury and cytoxicity, including apoptosis in vascular cells. Vascular endothelial cell apoptosis has been implicated in the pathophysiology and progression of atherosclerosis. We investigated whether the advanced glycation end-product inhibitor LR-90 could prevent MGO-induced apoptosis in human umbilical vascular endothelial cells (HUVECs). HUVECs were pre-treated with LR-90 and then stimulated with MGO. Cell morphology, cytotoxicity and apoptosis were evaluated by light microscopy, MTT assay, and Annexin V-FITC and propidium iodide double staining, respectively. Levels of Bax, Bcl-2, cytochrome c, mitogen-activated protein kinases (MAPKs) and caspase activities were assessed by Western blotting. Reactive oxygen species (ROS) generation and mitochondrial membrane potential (MMP) were measured with fluorescent probes. LR-90 dose-dependently prevented MGO-associated HUVEC cytotoxicity and apoptotic biochemical changes such as loss of MMP, increased Bax/Bcl-2 protein ratio, mitochondrial cytochrome c release and activation of caspase-3 and 9. Additionally, LR-90 blocked intracellular ROS formation and MAPK (p44/p42, p38, JNK) activation, though the latter seem to be not directly involved in MGO-induced HUVEC apoptosis. LR-90 prevents MGO-induced HUVEC apoptosis by inhibiting ROS and associated mitochondrial-dependent apoptotic signaling cascades, suggesting that LR-90 possess cytoprotective ability which could be beneficial in prevention of diabetic related-atherosclerosis.
Collapse
Affiliation(s)
- James L Figarola
- Departments of Diabetes and Metabolic Diseases Research, Beckman Research Institute of the City of Hope National Medical Center, NCI Designated Comprehensive Cancer Center, Gonda North, RM # 2108, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Cardiovascular disease, caused predominantly by atherosclerotic plaque rupture, remains one of the leading causes of death. However, the mechanism of plaque rupture remains largely unknown. Recent studies have linked high metabolic activity in inflamed atherosclerotic plaques to the development of plaque rupture. AGEs (advanced glycation end-products) are known to be formed as a result of high metabolic activity and are higher in rupture-prone than stable plaques. Furthermore, AGEs seem to be more than mere markers of metabolic activity, as recent studies have elucidated that AGEs and their major precursor, MG (methylglyoxal), may have an important role in the progression of atherosclerosis and plaque rupture. MG can be detoxified by Glo1 (glyoxalase I), thereby preventing the accumulation of MG and MG-derived AGEs. In the present review, data concerning MG, Glo1 and AGEs in the context of plaque phenotype are discussed.
Collapse
|
40
|
Sharma MK, Jalewa J, Hölscher C. Neuroprotective and anti-apoptotic effects of liraglutide on SH-SY5Y cells exposed to methylglyoxal stress. J Neurochem 2013; 128:459-71. [PMID: 24112036 DOI: 10.1111/jnc.12469] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 09/10/2013] [Accepted: 09/19/2013] [Indexed: 12/16/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) is a growth factor that has demonstrated neuroprotective properties in a range of studies. In an APPswe/PS1ΔE9 mouse model of Alzheimer's disease (AD), we previously found protective effects on memory formation, synaptic plasticity, synapse survival and a reduction of amyloid synthesis and plaque load in the brain. Here, we analyse the neuroprotective properties of the GLP-1 analogue liraglutide in human neuroblastoma cell line SH-SY5Y during methyl glyoxal stress. We show for the first time that cell viability was enhanced by liraglutide (XTT assay) in a dose-dependent way, while cytotoxicity (LDH assay) and apoptosis were reduced. Expression of the pro-survival Mcl1 signaling protein was increased, as was activation of cell survival kinases Akt, MEK1/2 and the transcription factor p90RSK. Liraglutide also decreased pro-apoptotic Bax and Bik expression. In addition, the membrane potential and the influx of calcium into the cell were enhanced by liraglutide. GLP-1 receptor expression was also increased by the drug. The results demonstrate a range of growth factor-related cytoprotective processes induced by liraglutide, which is currently on the market as a treatment for type 2 diabetes (Victoza®). It is also tested in clinical trials in patients with Alzheimer disease.
Collapse
Affiliation(s)
- Mohit K Sharma
- Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | | | | |
Collapse
|
41
|
van Eupen MGA, Schram MT, Colhoun HM, Scheijen JLJM, Stehouwer CDA, Schalkwijk CG. Plasma levels of advanced glycation endproducts are associated with type 1 diabetes and coronary artery calcification. Cardiovasc Diabetol 2013; 12:149. [PMID: 24134530 PMCID: PMC4015708 DOI: 10.1186/1475-2840-12-149] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/02/2013] [Indexed: 12/17/2022] Open
Abstract
Background Advanced glycation endproducts (AGEs) may play a role in the development of coronary artery calcification (CAC) in type 1 diabetes (T1DM). We studied plasma AGEs in association with T1DM and CAC, and whether or not the latter association could be explained by low-grade inflammation (LGI) or endothelial dysfunction (ED). Methods We studied 165 individuals with and 169 without T1DM. CAC was quantified in a CAC score based on CT-scanning. Plasma levels of protein-bound pentosidine, Nϵ-(carboxymethyl)lysine (CML) and Nϵ-(carboxyethyl)lysine (CEL) were measured with HPLC/UPLC with fluorescence detection or tandem-mass spectrometry. Tetrahydropyrimidine (THP) was measured with ELISA, as were HsCRP, and sVCAM-1 and vWF, as markers for LGI and ED, respectively. Associations were analyzed with ANCOVA and adjusted for age, sex, BMI, waist-to-hip ratio, smoking, blood pressure, lipid profile, eGFR and T1DM. Results Individuals with T1DM had higher plasma levels of pentosidine, CML and THP compared with controls; means (95% CI) were 0.69 (0.65-0.73) vs. 0.51 (0.48-0.54) nmol/mmol LYS, p < 0.001; 105 (102–107) vs. 93 (90–95) nmol/mmol LYS, p < 0.001; and 126 (118–134) vs. 113 (106–120) U/mL, p = 0.03, respectively. Levels of pentosidine were higher in individuals with T1DM with a moderate to high compared with a low CAC score, means (95% CI) were 0.81 (0.70-0.93) vs. 0.67 (0.63-0.71) nmol/mmol LYS, p = 0.03, respectively. This difference was not attenuated by adjustment for LGI or ED. Conclusions We found a positive association between pentosidine and CAC in T1DM. These results may indicate that AGEs are possibly involved in the development of CAC in individuals with T1DM.
Collapse
Affiliation(s)
| | | | | | | | | | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC) and Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, Maastricht 6200, MD, the Netherlands.
| |
Collapse
|