1
|
Prota G, Berloffa G, Awad W, Vacchini A, Chancellor A, Schaefer V, Constantin D, Littler DR, Colombo R, Nosi V, Mori L, Rossjohn J, De Libero G. Mitochondria regulate MR1 protein expression and produce self-metabolites that activate MR1-restricted T cells. Proc Natl Acad Sci U S A 2025; 122:e2418525122. [PMID: 40354545 DOI: 10.1073/pnas.2418525122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/11/2025] [Indexed: 05/14/2025] Open
Abstract
Mitochondria coordinate several metabolic pathways, producing metabolites that influence the immune response in various ways. It remains unclear whether mitochondria impact antigen presentation by the MHC-class-I-related antigen-presenting molecule, MR1, which presents small molecules to MR1-restricted T-lymphocytes. Here, we demonstrate that mitochondrial complex III and the enzyme dihydroorotate dehydrogenase are essential for the cell-surface expression of MR1 and for generating uridine- and thymidine-related compounds that bind to MR1 and are produced upon oxidation by reactive oxygen species. One mitochondria-derived immunogenic formylated metabolite we identified is 5-formyl-deoxyuridine (5-FdU). Structural studies indicate that 5-FdU binds in the A'-antigen-binding pocket of MR1, positioning the deoxyribose toward the surface of MR1 for TCR interaction. 5-FdU stimulates specific T cells and detects circulating T cells when loaded onto MR1-tetramers. 5-FdU-reactive cells resemble adaptive T cells and express the phenotypes of naïve, memory, and effector cells, indicating prior in vivo stimulation. These findings suggest that mitochondria may play a role in MR1-mediated immune surveillance.
Collapse
Affiliation(s)
- Gennaro Prota
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Giuliano Berloffa
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Wael Awad
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alessandro Vacchini
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Andrew Chancellor
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Verena Schaefer
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Daniel Constantin
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Dene R Littler
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Rodrigo Colombo
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Vladimir Nosi
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4YS, United Kingdom
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| |
Collapse
|
2
|
Berends E, Vangrieken P, Amiri N, van de Waarenburg MPH, Scheijen JLJM, Hermes DJHP, Wouters K, van Oostenbrugge RJ, Schalkwijk CG, Foulquier S. Increased Levels of Circulating Methylglyoxal Have No Consequence for Cerebral Microvascular Integrity and Cognitive Function in Young Healthy Mice. Mol Neurobiol 2025; 62:4190-4202. [PMID: 39414727 PMCID: PMC11880179 DOI: 10.1007/s12035-024-04552-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Diabetes and other age-related diseases are associated with an increased risk of cognitive impairment, but the underlying mechanisms remain poorly understood. Methylglyoxal (MGO), a by-product of glycolysis and a major precursor in the formation of advanced glycation end-products (AGEs), is increased in individuals with diabetes and other age-related diseases and is associated with microvascular dysfunction. We now investigated whether increased levels of circulating MGO can lead to cerebral microvascular dysfunction, blood-brain barrier (BBB) dysfunction, and cognitive impairment. Mice were supplemented or not with 50 mM MGO in drinking water for 13 weeks. Plasma and cortical MGO and MGO-derived AGEs were measured with UPLC-MS/MS. Peripheral and cerebral microvascular integrity and inflammation were investigated. Cerebral blood flow and neurovascular coupling were investigated with laser speckle contrast imaging, and cognitive tests were performed. We found a 2-fold increase in plasma MGO and an increase in MGO-derived AGEs in plasma and cortex. Increased plasma MGO did not lead to cerebral microvascular dysfunction, inflammation, or cognitive decline. This study shows that increased concentrations of plasma MGO are not associated with cerebral microvascular dysfunction and cognitive impairment in healthy mice. Future research should focus on the role of endogenously formed MGO in cognitive impairment.
Collapse
Affiliation(s)
- Eline Berends
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Philippe Vangrieken
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Naima Amiri
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Marjo P H van de Waarenburg
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Jean L J M Scheijen
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Denise J H P Hermes
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, the Netherlands
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands
| | - Kristiaan Wouters
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| | - Sébastien Foulquier
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands.
- Faculty of Health Medicine and Life Sciences, Department of Pharmacology and Toxicology, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
3
|
Rybnikářová A, Buchal R, Pláteník J. Ferritin with methylglyoxal produces reactive oxygen species but remains functional. Free Radic Res 2024; 58:702-722. [PMID: 39508662 DOI: 10.1080/10715762.2024.2417281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Iron is necessary for life, but the simultaneous iron-catalyzed formation of reactive oxygen species (ROS) is involved in pathogenesis of many diseases. One of them is diabetes mellitus, a widespread disease with severe long-term complications, including neuropathy, retinopathy, and nephropathy. Much evidence points to methylglyoxal, a potent glycating agent, as the key mediator of diabetic complications. In diabetes, there is also a peculiar dysregulation of iron homeostasis, leading to an expansion of redox-active iron. This in vitro study focuses on the interaction of methylglyoxal with ferritin, which is the main cellular protein for iron storage. Methylglyoxal effectively liberates iron from horse spleen ferritin, as well as synthetic iron cores; in both cases, it is partially mediated by superoxide. The interaction of methylglyoxal with ferritin increases the production of hydrogen peroxide, much above the generation of peroxide by methylglyoxal alone, in an iron-dependent manner. Glycation with methylglyoxal results in structural changes in ferritin. All of these findings can be demonstrated with pathophysiologically relevant (submillimolar) methylglyoxal concentrations. However, the rate of iron release by ascorbate, the ferroxidase activity, or the diameter of gated pores even in intensely glycated ferritin is not altered. In conclusion, although the functional features of ferritin resist alterations due to glycation, the interaction of methylglyoxal with ferritin liberates iron and markedly increases ROS production, both of which could enhance oxidative stress in vivo. Our findings may have implications for the pathogenesis of long-term diabetic complications, as well as for the use of ferritin as a nanocarrier in chemotherapy.
Collapse
Affiliation(s)
- Adriana Rybnikářová
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| | - Richard Buchal
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| | - Jan Pláteník
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| |
Collapse
|
4
|
Hellwig M, Diel P, Eisenbrand G, Grune T, Guth S, Henle T, Humpf HU, Joost HG, Marko D, Raupbach J, Roth A, Vieths S, Mally A. Dietary glycation compounds - implications for human health. Crit Rev Toxicol 2024; 54:485-617. [PMID: 39150724 DOI: 10.1080/10408444.2024.2362985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 08/17/2024]
Abstract
The term "glycation compounds" comprises a wide range of structurally diverse compounds that are formed endogenously and in food via the Maillard reaction, a chemical reaction between reducing sugars and amino acids. Glycation compounds produced endogenously are considered to contribute to a range of diseases. This has led to the hypothesis that glycation compounds present in food may also cause adverse effects and thus pose a nutritional risk to human health. In this work, the Senate Commission on Food Safety (SKLM) of the German Research Foundation (DFG) summarized data on formation, occurrence, exposure and toxicity of glycation compounds (Part A) and systematically assessed potential associations between dietary intake of defined glycation compounds and disease, including allergy, diabetes, cardiovascular and renal disease, gut/gastrotoxicity, brain/cognitive impairment and cancer (Part B). A systematic search in Pubmed (Medline), Scopus and Web of Science using a combination of keywords defining individual glycation compounds and relevant disease patterns linked to the subject area of food, nutrition and diet retrieved 253 original publications relevant to the research question. Of these, only 192 were found to comply with previously defined quality criteria and were thus considered suitable to assess potential health risks of dietary glycation compounds. For each adverse health effect considered in this assessment, however, only limited numbers of human, animal and in vitro studies were identified. While studies in humans were often limited due to small cohort size, short study duration, and confounders, experimental studies in animals that allow for controlled exposure to individual glycation compounds provided some evidence for impaired glucose tolerance, insulin resistance, cardiovascular effects and renal injury in response to oral exposure to dicarbonyl compounds, albeit at dose levels by far exceeding estimated human exposures. The overall database was generally inconsistent or inconclusive. Based on this systematic review, the SKLM concludes that there is at present no convincing evidence for a causal association between dietary intake of glycation compounds and adverse health effects.
Collapse
Affiliation(s)
- Michael Hellwig
- Chair of Special Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Patrick Diel
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | | | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Sabine Guth
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Thomas Henle
- Chair of Food Chemistry, TU Dresden, Dresden, Germany
| | | | - Hans-Georg Joost
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Jana Raupbach
- Institute of Food Chemistry, Technische Universität Braunschweig, Braunschweig, Germany
| | - Angelika Roth
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | | | - Angela Mally
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Ferreira-Junior MD, Cavalcante KVN, Costa JM, Bessa ASM, Amaro A, de Castro CH, Xavier CH, Silva S, Fonseca DA, Matafome P, Gomes RM. Early Methylglyoxal Exposure Leads to Worsened Cardiovascular Function in Young Rats. Nutrients 2024; 16:2029. [PMID: 38999777 PMCID: PMC11243563 DOI: 10.3390/nu16132029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Though maternal diabetes effects are well described in the literature, the effects of maternal diabetes in postnatal phases are often overlooked. Diabetic individuals have higher levels of circulating glycotoxins, and there is a positive correlation between maternal-derived glycotoxins and circulating glycotoxins in their progeny. Previous studies evaluated the metabolic effects of high glycotoxin exposure during lactation in adult animals. However, here we focus on the cardiovascular system of juvenile rats. METHODS For this, we used two experimental models: 1. High Methylglyoxal (MG) environment: pregnant Wistar rats were injected with PBS (VEH group) or Methylglyoxal (MG group; 60 mg/kg/day; orally, postnatal day (PND) 3 to PND14). 2. GLO-1 inhibition: pregnant Wistar rats were injected with dimethyl sulfoxide (VEH group) or a GLO-1 inhibitor (BBGC group; 5 mg/kg/day; subcutaneously, PND1-PND5). The offspring were evaluated at PND45. RESULTS MG offspring presented cardiac dysfunction and subtly worsened vasomotor responses in the presence of perivascular adipose tissue, without morphological alterations. In addition, an endogenous increase in maternal glycotoxins impacts offspring vasomotricity due to impaired redox status. CONCLUSIONS Our data suggest that early glycotoxin exposure led to cardiac and vascular impairments, which may increase the risk for developing cardiovascular diseases later in life.
Collapse
Affiliation(s)
- Marcos Divino Ferreira-Junior
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal (A.A.); (S.S.); (D.A.F.)
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| | - Keilah Valéria N. Cavalcante
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal (A.A.); (S.S.); (D.A.F.)
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| | - Jaqueline M. Costa
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| | - Amanda S. M. Bessa
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| | - Andreia Amaro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal (A.A.); (S.S.); (D.A.F.)
- Clinical and Academic Centre of Coimbra (CACC), 3004-531 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Carlos Henrique de Castro
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| | - Carlos Henrique Xavier
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| | - Sónia Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal (A.A.); (S.S.); (D.A.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Diogo A. Fonseca
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal (A.A.); (S.S.); (D.A.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal (A.A.); (S.S.); (D.A.F.)
- Clinical and Academic Centre of Coimbra (CACC), 3004-531 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Health School (ESTeSC), Polytechnic University of Coimbra, 3045-043 Coimbra, Portugal
| | - Rodrigo Mello Gomes
- Department of Physiological Sciences, Universidade Federal de Goiás, 74690-900 Goiás, Brazil; (J.M.C.); (A.S.M.B.); (C.H.d.C.); (C.H.X.); (R.M.G.)
| |
Collapse
|
6
|
Debras C, Cordova R, Mayén AL, Maasen K, Knaze V, Eussen SJPM, Schalkwijk CG, Huybrechts I, Tjønneland A, Halkjær J, Katzke V, Bajracharya R, Schulze MB, Masala G, Pala V, Pasanisi F, Macciotta A, Petrova D, Castañeda J, Santiuste C, Amiano P, Moreno-Iribas C, Borné Y, Sonestedt E, Johansson I, Esberg A, Aglago EK, Jenab M, Freisling H. Dietary intake of dicarbonyl compounds and changes in body weight over time in a large cohort of European adults. Br J Nutr 2024; 131:1902-1914. [PMID: 38383991 DOI: 10.1017/s0007114524000503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Dicarbonyl compounds are highly reactive precursors of advanced glycation end products (AGE), produced endogenously, present in certain foods and formed during food processing. AGE contribute to the development of adverse metabolic outcomes, but health effects of dietary dicarbonyls are largely unexplored. We investigated associations between three dietary dicarbonyl compounds, methylglyoxal (MGO), glyoxal (GO) and 3-deoxyglucosone (3-DG), and body weight changes in European adults. Dicarbonyl intakes were estimated using food composition database from 263 095 European Prospective Investigation into Cancer and Nutrition-Physical Activity, Nutrition, Alcohol, Cessation of Smoking, Eating Out of Home in Relation to Anthropometry participants with two body weight assessments (median follow-up time = 5·4 years). Associations between dicarbonyls and 5-year body-weight changes were estimated using mixed linear regression models. Stratified analyses by sex, age and baseline BMI were performed. Risk of becoming overweight/obese was assessed using multivariable-adjusted logistic regression. MGO intake was associated with 5-year body-weight gain of 0·089 kg (per 1-sd increase, 95 % CI 0·072, 0·107). 3-DG was inversely associated with body-weight change (-0·076 kg, -0·094, -0·058). No significant association was observed for GO (0·018 kg, -0·002, 0·037). In stratified analyses, GO was associated with body-weight gain among women and older participants (above median of 52·4 years). MGO was associated with higher body-weight gain among older participants. 3-DG was inversely associated with body-weight gain among younger and normal-weight participants. MGO was associated with a higher risk of becoming overweight/obese, while inverse associations were observed for 3-DG. No associations were observed for GO with overweight/obesity. Dietary dicarbonyls are inconsistently associated with body weight change among European adults. Further research is needed to clarify the role of these food components in overweight and obesity, their underlying mechanisms and potential public health implications.
Collapse
Affiliation(s)
- Charlotte Debras
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Reynalda Cordova
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Ana-Lucia Mayén
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Kim Maasen
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Viktoria Knaze
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM School for Cardiovascular Diseases/CAPHRI School for Public Health and Primary Care, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Inge Huybrechts
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Jytte Halkjær
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer research Center (DKFZ), Heidelberg, Germany
| | - Rashmita Bajracharya
- Department of Cancer Epidemiology, German Cancer research Center (DKFZ), Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Valeria Pala
- Epidemiology and Prevention Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Fabrizio Pasanisi
- Department of Clinical Medicine and Surgery School of Medicine, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Alessandra Macciotta
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Dafina Petrova
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029Madrid, Spain
| | - Jazmin Castañeda
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Carmen Santiuste
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, 2013 San Sebastian, Spain; Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, 20014 San Sebastián, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Conchi Moreno-Iribas
- Instituto de Salud Pública y Laboral de Navarra, 31003 Pamplona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain; Navarra Institute for Health Research (IdiSNA), 31008Pamplona, Spain
| | - Yan Borné
- Nutrition Epidemiology, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Emily Sonestedt
- Nutrition Epidemiology, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Anders Esberg
- Department of Odontology, Umeå University, Umeå, Sweden
| | - Elom Kouassivi Aglago
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Heinz Freisling
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| |
Collapse
|
7
|
Yang Y, Wang HL, Cheng RT, Zheng PR, Sun HP, Liu ZW, Yuan H, Liu XY, Gao WY, Li H. Determination of α-Dicarbonyl compounds in traditional Chinese herbal medicines. Fitoterapia 2024; 175:105928. [PMID: 38548027 DOI: 10.1016/j.fitote.2024.105928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/09/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
α-DCs (α-dicarbonyls) have been proven to be closely related to aging and the onset and development of many chronic diseases. The wide presence of this kind of components in various foods and beverages has been unambiguously determined, but their occurrence in various phytomedicines remains in obscurity. In this study, we established and evaluated an HPLC-UV method and used it to measure the contents of four α-DCs including 3-deoxyglucosone (3-DG), glyoxal (GO), methylglyoxal (MGO), and diacetyl (DA) in 35 Chinese herbs after they have been derivatized with 4-nitro-1,2-phenylenediamine. The results uncover that 3-DG is the major component among the α-DCs, being detectable in all the selected herbs in concentrations ranging from 22.80 μg/g in the seeds of Alpinia katsumadai to 7032.75 μg/g in the fruit of Siraitia grosuenorii. The contents of the other three compounds are much lower than those of 3-DG, with GO being up to 22.65 μg/g, MGO being up to 55.50 μg/g, and DA to 18.75 μg/g, respectively. The data show as well the contents of the total four α-DCs in the herbs are generally in a comparable level to those in various foods, implying that herb medicines may have potential risks on human heath in view of the α-DCs.
Collapse
Affiliation(s)
- Yang Yang
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; School of Pharmacy, Xi'an Medical University, 1 Xinwang Road, Xi'an, Shaanxi 710021, China
| | - Hai-Ling Wang
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Rui-Tong Cheng
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Pei-Rong Zheng
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Hui-Peng Sun
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Zhi-Wen Liu
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Heng Yuan
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Xue-Yi Liu
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Wen-Yun Gao
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| | - Heng Li
- College of Life Sciences and Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
8
|
Yang Y, Liu XY, Zhao Q, Wu D, Ren JT, Ma M, Li PY, Wu JC, Gao WY, Li H. Changes in α-Dicarbonyl Compound Contents during Storage of Various Fruits and Juices. Foods 2024; 13:1509. [PMID: 38790808 PMCID: PMC11119979 DOI: 10.3390/foods13101509] [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: 04/09/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
α-Dicarbonyl compounds (α-DCs) are commonly present in various foods. We conducted the investigation into concentration changes of α-DCs including 3-deoxyglucosone (3-DG), glyoxal (GO), and methylglyoxal (MGO) in fresh fruits and decapped commercial juices during storage at room temperature and 4 °C, as well as in homemade juices during storage at 4 °C. The studies indicate the presence of α-DCs in all samples. The initial contents of 3-DG in the commercial juices (6.74 to 65.61 μg/mL) are higher than those in the homemade ones (1.97 to 4.65 μg/mL) as well as fruits (1.58 to 3.33 μg/g). The initial concentrations of GO and MGO are normally less than 1 μg/mL in all samples. During storage, the α-DC levels in the fruits exhibit an initial increase followed by a subsequent decrease, whereas, in all juices, they tend to accumulate continuously over time. As expected, 4 °C storage reduces the increase rates of the α-DC concentrations in most samples. From the viewpoint of the α-DC contents, fruits and homemade juices should always be the first choice for daily intake of nutrients and commercial juices ought to be mostly avoided.
Collapse
Affiliation(s)
- Yang Yang
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
- School of Pharmacy, Xi’an Medical University, 1 Xinwang Road, Xi’an 710021, China
| | - Xue-Yi Liu
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Qian Zhao
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Dan Wu
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Jin-Tao Ren
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Meng Ma
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Pei-Yun Li
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Jia-Cai Wu
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Wen-Yun Gao
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| | - Heng Li
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (Y.Y.); (X.-Y.L.); (Q.Z.); (D.W.); (J.-T.R.); (M.M.); (P.-Y.L.); (J.-C.W.); (W.-Y.G.)
| |
Collapse
|
9
|
Oliveira AL, de Oliveira MG, Mónica FZ, Antunes E. Methylglyoxal and Advanced Glycation End Products (AGEs): Targets for the Prevention and Treatment of Diabetes-Associated Bladder Dysfunction? Biomedicines 2024; 12:939. [PMID: 38790901 PMCID: PMC11118115 DOI: 10.3390/biomedicines12050939] [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: 03/19/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024] Open
Abstract
Methylglyoxal (MGO) is a highly reactive α-dicarbonyl compound formed endogenously from 3-carbon glycolytic intermediates. Methylglyoxal accumulated in plasma and urine of hyperglycemic and diabetic individuals acts as a potent peptide glycation molecule, giving rise to advanced glycation end products (AGEs) like arginine-derived hydroimidazolone (MG-H1) and carboxyethyl-lysine (CEL). Methylglyoxal-derived AGEs exert their effects mostly via activation of RAGE, a cell surface receptor that initiates multiple intracellular signaling pathways, favoring a pro-oxidant environment through NADPH oxidase activation and generation of high levels of reactive oxygen species (ROS). Diabetic bladder dysfunction is a bothersome urological complication in patients with poorly controlled diabetes mellitus and may comprise overactive bladder, urge incontinence, poor emptying, dribbling, incomplete emptying of the bladder, and urinary retention. Preclinical models of type 1 and type 2 diabetes have further confirmed the relationship between diabetes and voiding dysfunction. Interestingly, healthy mice supplemented with MGO for prolonged periods exhibit in vivo and in vitro bladder dysfunction, which is accompanied by increased AGE formation and RAGE expression, as well as by ROS overproduction in bladder tissues. Drugs reported to scavenge MGO and to inactivate AGEs like metformin, polyphenols, and alagebrium (ALT-711) have shown favorable outcomes on bladder dysfunction in diabetic obese leptin-deficient and MGO-exposed mice. Therefore, MGO, AGEs, and RAGE levels may be critically involved in the pathogenesis of bladder dysfunction in diabetic individuals. However, there are no clinical trials designed to test drugs that selectively inhibit the MGO-AGEs-RAGE signaling, aiming to reduce the manifestations of diabetes-associated bladder dysfunction. This review summarizes the current literature on the role of MGO-AGEs-RAGE-ROS axis in diabetes-associated bladder dysfunction. Drugs that directly inactivate MGO and ameliorate bladder dysfunction are also reviewed here.
Collapse
Affiliation(s)
| | | | | | - Edson Antunes
- Department of Translational Medicine, Pharmacology Area, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (A.L.O.); (M.G.d.O.); (F.Z.M.)
| |
Collapse
|
10
|
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
|
11
|
Kim Y. Blood and Tissue Advanced Glycation End Products as Determinants of Cardiometabolic Disorders Focusing on Human Studies. Nutrients 2023; 15:nu15082002. [PMID: 37111220 PMCID: PMC10144557 DOI: 10.3390/nu15082002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiometabolic disorders are characterised by a cluster of interactive risk determinants such as increases in blood glucose, lipids and body weight, as well as elevated inflammation and oxidative stress and gut microbiome changes. These disorders are associated with onset of type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). T2DM is strongly associated with CVD. Dietary advanced glycation end products (dAGEs) attributable from modern diets high in sugar and/or fat, highly processed foods and high heat-treated foods can contribute to metabolic etiologies of cardiometabolic disorders. This mini review aims to determine whether blood dAGEs levels and tissue dAGEs levels are determinants of the prevalence of cardiometabolic disorders through recent human studies. ELISA (enzyme-linked immunosorbent assay), high-performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS) for blood dAGEs measurement and skin auto fluorescence (SAF) for skin AGEs measurement can be used. Recent human studies support that a diet high in AGEs can negatively influence glucose control, body weight, blood lipid levels and vascular health through the elevated oxidative stress, inflammation, blood pressure and endothelial dysfunction compared with a diet low in AGEs. Limited human studies suggested a diet high in AGEs could negatively alter gut microbiota. SAF could be considered as one of the predictors affecting risks for cardiometabolic disorders. More intervention studies are needed to determine how dAGEs are associated with the prevalence of cardiometabolic disorders through gut microbiota changes. Further human studies are conducted to find the association between CVD events, CVD mortality and total mortality through SAF measurement, and a consensus on whether tissue dAGEs act as a predictor of CVD is required.
Collapse
Affiliation(s)
- Yoona Kim
- Department of Food and Nutrition, Institute of Agriculture and Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
12
|
Hanssen NMJ, Tikellis C, Pickering RJ, Dragoljevic D, Lee MKS, Block T, Scheijen JL, Wouters K, Miyata T, Cooper ME, Murphy AJ, Thomas MC, Schalkwijk CG. Pyridoxamine prevents increased atherosclerosis by intermittent methylglyoxal spikes in the aortic arches of ApoE -/- mice. Biomed Pharmacother 2023; 158:114211. [PMID: 36916437 DOI: 10.1016/j.biopha.2022.114211] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Methylglyoxal (MGO) is a reactive glucose metabolite linked to diabetic cardiovascular disease (CVD). MGO levels surge during intermittent hyperglycemia. We hypothesize that these MGO spikes contribute to atherosclerosis, and that pyridoxamine as a MGO quencher prevents this injury. To study this, we intravenously injected normoglycemic 8-week old male C57Bl6 ApoE-/- mice with normal saline (NS, n = 10) or 25 µg MGO for 10 consecutive weeks (MGOiv, n = 11) with or without 1 g/L pyridoxamine (MGOiv+PD, n = 11) in the drinking water. We measured circulating immune cells by flow cytometry. We quantified aortic arch lesion area in aortic roots after Sudan-black staining. We quantified the expression of inflammatory genes in the aorta by qPCR. Intermittent MGO spikes weekly increased atherosclerotic burden in the arch 1.8-fold (NS: 0.9 ± 0.1 vs 1.6 ± 0.2 %), and this was prevented by pyridoxamine (0.8 ± 0.1 %). MGOiv spikes increased circulating neutrophils and monocytes (2-fold relative to NS) and the expression of ICAM (3-fold), RAGE (5-fold), S100A9 (2-fold) and MCP1 (2-fold). All these changes were attenuated by pyridoxamine. This study suggests that MGO spikes damages the vasculature independently of plasma glucose levels. Pyridoxamine and potentially other approaches to reduce MGO may prevent excess cardiovascular risk in diabetes.
Collapse
Affiliation(s)
- Nordin M J Hanssen
- Amsterdam Diabetes Centrum, Internal and Vascular Medicine, Amsterdam University Medical Centres, location AMC, Amsterdam, the Netherlands
| | - Chris Tikellis
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Raelene J Pickering
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Dragana Dragoljevic
- Dept. of leukocyte biology and haematopoiesis, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Man Kit Sam Lee
- Dept. of leukocyte biology and haematopoiesis, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Tomasz Block
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jean Ljm Scheijen
- Dept. of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Kristiaan Wouters
- Dept. of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Toshio Miyata
- Division of Molecular Medicine and Therapy, Tohoku University Graduate School of Medicine, Japan
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Dept. of leukocyte biology and haematopoiesis, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Merlin C Thomas
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Casper G Schalkwijk
- Dept. of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands.
| |
Collapse
|
13
|
Van den Eynde MDG, Houben AJHM, Scheijen JLJM, Linkens AMA, Niessen PM, Simons N, Hanssen NMJ, Kusters YHAM, Eussen SJMP, Miyata T, Stehouwer CDA, Schalkwijk CG. Pyridoxamine reduces methylglyoxal and markers of glycation and endothelial dysfunction, but does not improve insulin sensitivity or vascular function in abdominally obese individuals: A randomized double-blind placebo-controlled trial. Diabetes Obes Metab 2023; 25:1280-1291. [PMID: 36655410 DOI: 10.1111/dom.14977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
AIM To investigate the effects of pyridoxamine (PM), a B6 vitamer and dicarbonyl scavenger, on glycation and a large panel of metabolic and vascular measurements in a randomized double-blind placebo-controlled trial in abdominally obese individuals. MATERIALS AND METHODS Individuals (54% female; mean age 50 years; mean body mass index 32 kg/m2 ) were randomized to an 8-week intervention with either placebo (n = 36), 25 mg PM (n = 36) or 200 mg PM (n = 36). We assessed insulin sensitivity, β-cell function, insulin-mediated microvascular recruitment, skin microvascular function, flow-mediated dilation, and plasma inflammation and endothelial function markers. PM metabolites, dicarbonyls and advanced glycation endproducts (AGEs) were measured using ultra-performance liquid chromatography tandem mass spectrometry. Treatment effects were evaluated by one-way ANCOVA. RESULTS In the high PM dose group, we found a reduction of plasma methylglyoxal (MGO) and protein-bound Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1), as compared to placebo. We found a reduction of the endothelial dysfunction marker soluble vascular cell adhesion molecule-1 (sVCAM-1) in the low and high PM dose group and of soluble intercellular adhesion molecule-1 (sICAM-1) in the high PM dose, as compared to placebo. We found no treatment effects on insulin sensitivity, vascular function or other functional outcome measurements. CONCLUSIONS This study shows that PM is metabolically active and reduces MGO, AGEs, sVCAM-1 and sICAM-1, but does not affect insulin sensitivity and vascular function in abdominally obese individuals. The reduction in adhesion markers is promising because these are important in the pathogenesis of endothelial damage and atherosclerosis.
Collapse
Affiliation(s)
- Mathias D G Van den Eynde
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Alfons J H M Houben
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Jean L J M Scheijen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Armand M A Linkens
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Petra M Niessen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Nynke Simons
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Nordin M J Hanssen
- Amsterdam Diabetes Center, Department of Internal and Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Yvo H A M Kusters
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Simone J M P Eussen
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht, The Netherlands
| | - Toshio Miyata
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| |
Collapse
|
14
|
Kolibabka M, Dannehl M, Oezer K, Murillo K, Huang H, Riemann S, Hoffmann S, Gretz N, Schlotterer A, Feng Y, Hammes HP. Differences in junction-associated gene expression changes in three rat models of diabetic retinopathy with similar neurovascular phenotype. Neurobiol Dis 2023; 176:105961. [PMID: 36526091 DOI: 10.1016/j.nbd.2022.105961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/24/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Diabetic retinopathy, also defined as microvascular complication of diabetes mellitus, affects the entire neurovascular unit with specific aberrations in every compartment. Neurodegeneration, glial activation and vasoregression are observed consistently in models of diabetic retinopathy. However, the order and the severity of these aberrations varies in different models, which is also true in patients. In this study, we analysed rat models of diabetic retinopathy with similar phenotypes to identify key differences in the pathogenesis. For this, we focussed on intercellular junction-associated gene expression, which are important for the communication and homeostasis within the neurovascular unit. Streptozotocin-injected diabetic Wistar rats, methylglyoxal supplemented Wistar rats and polycystin-2 transgenic (PKD) rats were analysed for neuroretinal function, vasoregression and retinal expression of junction-associated proteins. In all three models, neuroretinal impairment and vasoregression were observed, but gene expression profiling of junction-associated proteins demonstrated nearly no overlap between the three models. However, the differently expressed genes were from the main classes of claudins, connexins and integrins in all models. Changes in Rcor1 expression in diabetic rats and Egr1 expression in PKD rats confirmed the differences in upstream transcription factor level between the models. In PKD rats, a possible role for miRNA regulation was observed, indicated by an upregulation of miR-26b-5p, miR-122-5p and miR-300-3p, which was not observed in the other models. In silico allocation of connexins revealed not only differences in regulated subtypes, but also in affected retinal cell types, as well as connexin specific upstream regulators Sox7 and miR-92a-3p. In this study, we demonstrate that, despite their similar phenotype, models for diabetic retinopathy exhibit significant differences in their pathogenic pathways and primarily affected cell types. These results underline the importance for more sensitive diagnostic tools to identify pathogenic clusters in patients as the next step towards a desperately needed personalized therapy.
Collapse
Affiliation(s)
- Matthias Kolibabka
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; European Center for Angioscience, Ludolf-Krehl-Strasse 13 - 17, 68167 Mannheim, Germany.
| | - Marcus Dannehl
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; European Center for Angioscience, Ludolf-Krehl-Strasse 13 - 17, 68167 Mannheim, Germany
| | - Kübra Oezer
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Katharina Murillo
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; European Center for Angioscience, Ludolf-Krehl-Strasse 13 - 17, 68167 Mannheim, Germany
| | - Hongpeng Huang
- Experimental Pharmacology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Strasse 13 - 17, 68167 Manheim, Germany
| | - Sarah Riemann
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; European Center for Angioscience, Ludolf-Krehl-Strasse 13 - 17, 68167 Mannheim, Germany
| | - Sigrid Hoffmann
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Andrea Schlotterer
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; European Center for Angioscience, Ludolf-Krehl-Strasse 13 - 17, 68167 Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Strasse 13 - 17, 68167 Manheim, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; European Center for Angioscience, Ludolf-Krehl-Strasse 13 - 17, 68167 Mannheim, Germany
| |
Collapse
|
15
|
Schalkwijk CG, Micali LR, Wouters K. Advanced glycation endproducts in diabetes-related macrovascular complications: focus on methylglyoxal. Trends Endocrinol Metab 2023; 34:49-60. [PMID: 36446668 DOI: 10.1016/j.tem.2022.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022]
Abstract
Diabetes is associated with vascular injury and the onset of macrovascular complications. Advanced glycation endproducts (AGEs) and the AGE precursor methylglyoxal (MGO) have been identified as key players in establishing the relationship between diabetes and vascular injury. While most research has focused on the link between AGEs and vascular injury, less is known about the effects of MGO on vasculature. In this review, we focus on the mechanisms linking AGEs and MGO to the development of atherosclerosis. AGEs and MGO are involved in many stages of atherosclerosis progression. However, more research is needed to determine the exact mechanisms underlying these effects. Nevertheless, AGEs and MGO could represent valid therapeutic targets for the macrovascular complications of diabetes.
Collapse
Affiliation(s)
- Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, MUMC+, Maastricht, The Netherlands
| | | | - Kristiaan Wouters
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, MUMC+, Maastricht, The Netherlands.
| |
Collapse
|
16
|
Maasen K, Eussen SJPM, Dagnelie PC, Houben AJHM, Webers CAB, Schram MT, Berendschot TTJM, Stehouwer CDA, Opperhuizen A, van Greevenbroek MMJ, Schalkwijk CG. Habitual intake of dietary methylglyoxal is associated with less low-grade inflammation: the Maastricht Study. Am J Clin Nutr 2022; 116:1715-1728. [PMID: 36055771 PMCID: PMC9761753 DOI: 10.1093/ajcn/nqac195] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 07/08/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Dicarbonyls are major reactive precursors of advanced glycation endproducts (AGEs). Dicarbonyls are formed endogenously and also during food processing. Circulating dicarbonyls and AGEs are associated with inflammation and microvascular complications of diabetes, but for dicarbonyls from the diet these associations are currently unknown. OBJECTIVES We sought to examine the associations of dietary dicarbonyl intake with low-grade inflammation and microvascular function. METHODS In 2792 participants (mean ± SD age: 60 ± 8 y; 50% men; 26% type 2 diabetes) of the population-based cohort the Maastricht Study, we estimated the habitual intake of the dicarbonyls methylglyoxal (MGO), glyoxal (GO), and 3-deoxyglucosone (3-DG) by linking FFQ outcome data to our food composition database of the MGO, GO, and 3-DG content of >200 foods. Low-grade inflammation was assessed as six plasma biomarkers, which were compiled in a z score. Microvascular function was assessed as four plasma biomarkers, compiled in a zscore; as diameters and flicker light-induced dilation in retinal microvessels; as heat-induced skin hyperemic response; and as urinary albumin excretion. Cross-sectional associations of dietary dicarbonyls with low-grade inflammation and microvascular function were investigated using linear regression with adjustments for age, sex, potential confounders related to cardiometabolic risk factors, and lifestyle and dietary factors. RESULTS Fully adjusted analyses revealed that higher intake of MGO was associated with a lower z score for inflammation [standardized β coefficient (STD β): -0.05; 95% CI: -0.09 to -0.01, with strongest inverse associations for hsCRP and TNF-α: both -0.05; -0.10 to -0.01]. In contrast, higher dietary MGO intake was associated with impaired retinal venular dilation after full adjustment (STD β: -0.07; 95% CI: -0.12 to -0.01), but not with the other features of microvascular function. GO and 3-DG intakes were not consistently associated with any of the outcomes. CONCLUSION Higher habitual intake of MGO was associated with less low-grade inflammation. This novel, presumably beneficial, association is the first observation of an association between MGO intake and health outcomes in humans and warrants further investigation.
Collapse
Affiliation(s)
- Kim Maasen
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute/CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pieter C Dagnelie
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute/CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Alfons J H M Houben
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Miranda T Schram
- Department of Internal Medicine, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Tos T J M Berendschot
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Antoon Opperhuizen
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
- Office for Risk Assessment and Research, Netherlands Food and Consumer Product Safety Authority, Utrecht, The Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
17
|
Methylglyoxal and glyoxalase 1-a metabolic stress pathway-linking hyperglycemia to the unfolded protein response and vascular complications of diabetes. Clin Sci (Lond) 2022; 136:819-824. [PMID: 35635155 PMCID: PMC9152679 DOI: 10.1042/cs20220099] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022]
Abstract
The study of the glyoxalase system by Thornalley and co-workers in clinical diabetes mellitus and correlation with diabetic complications revealed increased exposure of patients with diabetes to the reactive, dicarbonyl metabolite methylglyoxal (MG). Twenty-eight years later, extended and built on by Thornalley and co-workers and others, the glyoxalase system is an important pathway contributing to the development of insulin resistance and vascular complications of diabetes. Other related advances have been: characterization of a new kind of metabolic stress—‘dicarbonyl stress’; identification of the major physiological advanced glycation endproduct (AGE), MG-H1; physiological substrates of the unfolded protein response (UPR); new therapeutic agents—‘glyoxalase 1 (Glo1) inducers’; and a refined mechanism underlying the link of dysglycemia to the development of insulin resistance and vascular complications of diabetes.
Collapse
|
18
|
Emerging Glycation-Based Therapeutics-Glyoxalase 1 Inducers and Glyoxalase 1 Inhibitors. Int J Mol Sci 2022; 23:ijms23052453. [PMID: 35269594 PMCID: PMC8910005 DOI: 10.3390/ijms23052453] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
The abnormal accumulation of methylglyoxal (MG) leading to increased glycation of protein and DNA has emerged as an important metabolic stress, dicarbonyl stress, linked to aging, and disease. Increased MG glycation produces inactivation and misfolding of proteins, cell dysfunction, activation of the unfolded protein response, and related low-grade inflammation. Glycation of DNA and the spliceosome contribute to an antiproliferative and apoptotic response of high, cytotoxic levels of MG. Glyoxalase 1 (Glo1) of the glyoxalase system has a major role in the metabolism of MG. Small molecule inducers of Glo1, Glo1 inducers, have been developed to alleviate dicarbonyl stress as a prospective treatment for the prevention and early-stage reversal of type 2 diabetes and prevention of vascular complications of diabetes. The first clinical trial with the Glo1 inducer, trans-resveratrol and hesperetin combination (tRES-HESP)-a randomized, double-blind, placebo-controlled crossover phase 2A study for correction of insulin resistance in overweight and obese subjects, was completed successfully. tRES-HESP corrected insulin resistance, improved dysglycemia, and low-grade inflammation. Cell permeable Glo1 inhibitor prodrugs have been developed to induce severe dicarbonyl stress as a prospective treatment for cancer-particularly for high Glo1 expressing-related multidrug-resistant tumors. The prototype Glo1 inhibitor is prodrug S-p-bromobenzylglutathione cyclopentyl diester (BBGD). It has antitumor activity in vitro and in tumor-bearing mice in vivo. In the National Cancer Institute human tumor cell line screen, BBGD was most active against the glioblastoma SNB-19 cell line. Recently, potent antitumor activity was found in glioblastoma multiforme tumor-bearing mice. High Glo1 expression is a negative survival factor in chemotherapy of breast cancer where adjunct therapy with a Glo1 inhibitor may improve treatment outcomes. BBGD has not yet been evaluated clinically. Glycation by MG now appears to be a pathogenic process that may be pharmacologically manipulated for therapeutic outcomes of potentially important clinical impact.
Collapse
|
19
|
Methylglyoxal Scavengers Attenuate Angiogenesis Dysfunction Induced by Methylglyoxal and Oxygen-Glucose Deprivation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8854457. [PMID: 35035668 PMCID: PMC8754597 DOI: 10.1155/2022/8854457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 12/11/2022]
Abstract
Cerebral endothelial cells play an essential role in brain angiogenesis, and their function has been found to be impaired in diabetes. Methylglyoxal (MG) is a highly reactive dicarbonyl metabolite of glucose formed mainly during glycolysis, and its levels can be elevated in hyperglycemic conditions. MG is a potent precursor of AGEs (advanced glycation end-products). In this study, we investigated if MG can induce angiogenesis dysfunction and whether MG scavengers can ameliorate angiogenesis dysfunction induced by MG. Here, we used cultured human brain microvascular endothelial cells (HBMECs) treated with MG and oxygen-glucose deprivation (OGD) to mimic diabetic stroke in vitro. We also used the MG challenged chicken embryo chorioallantoic membrane (CAM) to study angiogenesis in vivo. Interestingly, administration of MG significantly impaired cell proliferation, cell migration, and tube formation and decreased protein expression of angiogenesis-related factors, which was rescued by three different MG scavengers, glyoxalase 1 (GLO1), aminoguanidine (AG), and N-acetyl cysteine (NAC). In cultured CAM, MG exposure significantly reduced angiogenesis and the angiogenesis-related dysfunction could be attenuated by pretreatment with AG or NAC. Treatment of cultured HBMECs with MG plus OGD increased cellular apoptosis significantly, which could be prevented by exposure to GLO1, AG, or NAC. We also noted that administration of MG increased cellular oxidative stress as measured by reactive oxygen species (ROS) generation, enhanced AGE accumulation, and receptor for advanced glycation end-product (RAGE) expression in the cultured HBMECs, which were partially reversed by GLO1, AG, or NAC. Taken together, our findings demonstrated that GLO1, AG, or NAC administration can ameliorate MG-induced angiogenesis dysfunction, and this can be mainly attributed to attenuated ROS production, reduced cellular apoptosis, and increased levels of angiogenic factors. Overall, this study suggested that GLO1, AG, or NAC may be promising candidate compounds for the treatment of angiogenesis dysfunction caused by hyperglycemia in diabetic ischemic stroke.
Collapse
|
20
|
Zhao Y, Tang Y, Sang S. Dietary Quercetin Reduces Plasma and Tissue Methylglyoxal and Advanced Glycation End Products in Healthy Mice Treated with Methylglyoxal. J Nutr 2021; 151:2601-2609. [PMID: 34091674 DOI: 10.1093/jn/nxab176] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/15/2021] [Accepted: 05/11/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Methylglyoxal (MGO), a precursor of advanced glycation end products (AGEs), has been linked to AGEs-associated diseases. OBJECTIVES This study investigated the efficacy and mechanisms of dietary quercetin in decreasing plasma and tissue concentrations of MGO and AGEs in MGO-administered mice. METHODS Male, 6-wk-old CD-1 mice were administered AIN-93G diet and water (Con) or 0.12% MGO in water (MGO) or MGO plus 0.2% (0.2Q) dietary quercetin for 1 wk (n = 5) (experiment 1), and water (Con), 0.12% MGO (MGO), or MGO plus 0.1% (0.1Q), 0.2% (0.2Q), or 0.4% (0.4Q) dietary quercetin for 6 wk (n = 10) (experiment 2). The plasma, kidney, and liver concentrations of MGO, quercetin, and isorhamnetin and their trapping adducts with MGO were determined by LC-MS, and AGE concentrations were measured by the fluorescent method. Furthermore, the expressions of glyoxalase I/II (GLO I/II) and aldose reductase (AR), MGO detoxification enzymes, were determined by Western blot. One-factor ANOVA and post hoc Dunnett's or Tukey's test were used to analyze the data. RESULTS After 1 wk of treatment, the MGO concentrations in plasma (20.2%) and kidney (29.9%) in 0.2Q mice were significantly lower than those in MGO mice. After 6 wk of treatment, the concentrations of MGO in the plasma (14.7-18.6%), kidney (20-20.8%), liver (15.4-18.6%), and tissue AGEs (28-36.8%) in 0.1Q, 0.2Q, and 0.4Q mice were significantly lower than those in MGO mice. The plasma concentrations of quercetin, isorhamnetin, and their MGO adducts were dose-dependently increased after quercetin administration. In addition, after 6 wk of quercetin administration, the expressions of GLO I/II and AR in the liver and kidney were significantly upregulated to promote MGO detoxification compared with MGO-treated mice. CONCLUSIONS Quercetin reduced plasma and tissue MGO concentrations and inhibited AGE formation by trapping MGO and regulating the MGO detoxification systems in MGO-administered healthy mice.
Collapse
Affiliation(s)
- Yantao Zhao
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, NC, USA
| | - Yao Tang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, NC, USA
| | - Shengmin Sang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, NC, USA
| |
Collapse
|
21
|
Effect of Advanced Glycation End-Products and Excessive Calorie Intake on Diet-Induced Chronic Low-Grade Inflammation Biomarkers in Murine Models. Nutrients 2021; 13:nu13093091. [PMID: 34578967 PMCID: PMC8468789 DOI: 10.3390/nu13093091] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022] Open
Abstract
Chronic Low-Grade Inflammation (CLGI) is a non-overt inflammatory state characterized by a continuous activation of inflammation mediators associated with metabolic diseases. It has been linked to the overconsumption of Advanced Glycation End-Products (AGEs), and/or macronutrients which lead to an increase in local and systemic pro-inflammatory biomarkers in humans and animal models. This review provides a summary of research into biomarkers of diet-induced CLGI in murine models, with a focus on AGEs and obesogenic diets, and presents the physiological effects described in the literature. Diet-induced CLGI is associated with metabolic endotoxemia, and/or gut microbiota remodeling in rodents. The mechanisms identified so far are centered on pro-inflammatory axes such as the interaction between AGEs and their main receptor AGEs (RAGE) or increased levels of lipopolysaccharide. The use of murine models has helped to elucidate the local and systemic expression of CLGI mediators. These models have enabled significant advances in identification of diet-induced CLGI biomarkers and resultant physiological effects. Some limitations on the translational (murine → humans) use of biomarkers may arise, but murine models have greatly facilitated the testing of specific dietary components. However, there remains a lack of information at the whole-organism level of organization, as well as a lack of consensus on the best biomarker for use in CLGI studies and recommendations as to future research conclude this review.
Collapse
|
22
|
Wouters K, Cento AS, Gaens KH, Teunissen M, Scheijen JLJM, Barutta F, Chiazza F, Collotta D, Aragno M, Gruden G, Collino M, Schalkwijk CG, Mastrocola R. Deletion of RAGE fails to prevent hepatosteatosis in obese mice due to impairment of other AGEs receptors and detoxifying systems. Sci Rep 2021; 11:17373. [PMID: 34462492 PMCID: PMC8405685 DOI: 10.1038/s41598-021-96859-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Advanced glycation endproducts (AGEs) are involved in several diseases, including NAFLD and NASH. RAGE is the main receptor mediating the pro-inflammatory signalling induced by AGEs. Therefore, targeting of RAGE has been proposed for prevention of chronic inflammatory diseases. However, the role of RAGE in the development of NAFLD and NASH remains poorly understood. We thus aimed to analyse the effect of obesity on AGEs accumulation, AGE-receptors and AGE-detoxification, and whether the absence of RAGE might improve hepatosteatosis and inflammation, by comparing the liver of lean control, obese (LeptrDb-/-) and obese RAGE-deficient (RAGE-/- LeptrDb-/-) mice. Obesity induced AGEs accumulation and RAGE expression with hepatosteatosis and inflammation in LeptrDb-/-, compared to lean controls. Despite the genetic deletion of RAGE in the LeptrDb-/- mice, high levels of intrahepatic AGEs were maintained accompanied by decreased expression of the protective AGE-receptor-1, impaired AGE-detoxifying system glyoxalase-1, and increased expression of the alternative AGE-receptor galectin-3. We also found sustained hepatosteatosis and inflammation as determined by persistent activation of the lipogenic SREBP1c and proinflammatory NLRP3 signalling pathways. Thus, RAGE targeting is not effective in the prevention of NAFLD in conditions of obesity, likely due to the direct liver specific crosstalk of RAGE with other AGE-receptors and AGE-detoxifying systems.
Collapse
Affiliation(s)
- Kristiaan Wouters
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Alessia S. Cento
- grid.7605.40000 0001 2336 6580Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Katrien H. Gaens
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Margee Teunissen
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands
| | - Jean L. J. M. Scheijen
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Federica Barutta
- grid.7605.40000 0001 2336 6580Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fausto Chiazza
- grid.16563.370000000121663741Department of Drug Sciences, University of Eastern Piedmont, Novara, Italy
| | - Debora Collotta
- grid.7605.40000 0001 2336 6580Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Manuela Aragno
- grid.7605.40000 0001 2336 6580Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Gabriella Gruden
- grid.7605.40000 0001 2336 6580Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Collino
- grid.7605.40000 0001 2336 6580Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Casper G. Schalkwijk
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Raffaella Mastrocola
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.7605.40000 0001 2336 6580Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| |
Collapse
|
23
|
Wang W, Chen J, Ma H, Xing W, Lv N, Zhang B, Xu H, Wang W, Lou K. An "AND"-logic-gate-based fluorescent probe with dual reactive sites for monitoring extracellular methylglyoxal level changes of activated macrophages. Chem Commun (Camb) 2021; 57:8166-8169. [PMID: 34318802 DOI: 10.1039/d1cc01859k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
An "AND"-logic-gate-based fluorescent probe NAP-DCP-4 with dual reactive sites is reported, which has improved selectivity for methylglyoxal over glyoxal, featuring formaldehyde-enhanced methylglyoxal detection and irreversible and reversible turn-on fluorescence responses at different excitation wavelengths. Its cell-impermeability enables facile monitoring of extracellular methylglyoxal level changes in the supernatant of activated macrophages.
Collapse
Affiliation(s)
- Wenli Wang
- State Key Laboratory of Bioengineering Reactor, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
The Glyoxalase System in Age-Related Diseases: Nutritional Intervention as Anti-Ageing Strategy. Cells 2021; 10:cells10081852. [PMID: 34440621 PMCID: PMC8393707 DOI: 10.3390/cells10081852] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/19/2022] Open
Abstract
The glyoxalase system is critical for the detoxification of advanced glycation end-products (AGEs). AGEs are toxic compounds resulting from the non-enzymatic modification of biomolecules by sugars or their metabolites through a process called glycation. AGEs have adverse effects on many tissues, playing a pathogenic role in the progression of molecular and cellular aging. Due to the age-related decline in different anti-AGE mechanisms, including detoxifying mechanisms and proteolytic capacities, glycated biomolecules are accumulated during normal aging in our body in a tissue-dependent manner. Viewed in this way, anti-AGE detoxifying systems are proposed as therapeutic targets to fight pathological dysfunction associated with AGE accumulation and cytotoxicity. Here, we summarize the current state of knowledge related to the protective mechanisms against glycative stress, with a special emphasis on the glyoxalase system as the primary mechanism for detoxifying the reactive intermediates of glycation. This review focuses on glyoxalase 1 (GLO1), the first enzyme of the glyoxalase system, and the rate-limiting enzyme of this catalytic process. Although GLO1 is ubiquitously expressed, protein levels and activities are regulated in a tissue-dependent manner. We provide a comparative analysis of GLO1 protein in different tissues. Our findings indicate a role for the glyoxalase system in homeostasis in the eye retina, a highly oxygenated tissue with rapid protein turnover. We also describe modulation of the glyoxalase system as a therapeutic target to delay the development of age-related diseases and summarize the literature that describes the current knowledge about nutritional compounds with properties to modulate the glyoxalase system.
Collapse
|
25
|
Systemic inflammation down-regulates glyoxalase-1 expression: an experimental study in healthy males. Biosci Rep 2021; 41:229081. [PMID: 34156474 PMCID: PMC8411911 DOI: 10.1042/bsr20210954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Hypoxia and inflammation are hallmarks of critical illness, related
to multiple organ failure. A possible mechanism leading to multiple organ
failure is hypoxia- or inflammation-induced down-regulation of the detoxifying
glyoxalase system that clears dicarbonyl stress. The dicarbonyl methylglyoxal
(MGO) is a highly reactive agent produced by metabolic pathways such as
anaerobic glycolysis and gluconeogenesis. MGO leads to protein damage and
ultimately multi-organ failure. Whether detoxification of MGO into D-lactate by
glyoxalase functions appropriately under conditions of hypoxia and inflammation
is largely unknown. We investigated the effect of inflammation and hypoxia on
the MGO pathway in humans in vivo. Methods: After prehydration with glucose 2.5% solution, ten healthy males
were exposed to hypoxia (arterial saturation 80–85%) for 3.5 h
using an air-tight respiratory helmet, ten males to experimental endotoxemia
(LPS 2 ng/kg i.v.), ten males to LPS+hypoxia and ten males to none of these
interventions (control group). Serial blood samples were drawn, and glyoxalase-1
mRNA expression, MGO, methylglyoxal-derived hydroimidazolone-1 (MG-H1),
D-lactate and L-lactate levels, were measured serially. Results: Glyoxalase-1 mRNA expression decreased in the LPS (β
(95%CI); -0.87 (-1.24; -0.50) and the LPS+hypoxia groups; -0.78 (-1.07;
-0.48) (P<0.001). MGO was equal between groups, whereas
MG-H1 increased over time in the control group only
(P=0.003). D-Lactate was increased in all four groups.
L-Lactate was increased in all groups, except in the control group. Conclusion: Systemic inflammation downregulates glyoxalase-1 mRNA expression in
humans. This is a possible mechanism leading to cell damage and multi-organ
failure in critical illness with potential for intervention.
Collapse
|
26
|
Bora S, Shankarrao Adole P. Carbonyl stress in diabetics with acute coronary syndrome. Clin Chim Acta 2021; 520:78-86. [PMID: 34090879 DOI: 10.1016/j.cca.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 01/17/2023]
Abstract
The prevalence and incidence of diabetes mellitus (DM) are increasing worldwide bringing with it a significantly higher rate of complications. Various mechanisms such as carbonyl stress, polyol pathway, oxidative stress, hexosamine pathways, diacylglycerol/protein kinase-C activation, etc., are responsible for the pathogenesis of DM and its complications. Persistent hyperglycaemia and inhibition of metabolising and detoxifying enzymes lead to the excessive synthesis of carbonyl compounds such as methylglyoxal, glyoxal, and 3-deoxyglucosone, resulting in carbonyl stress. The substrates, metabolizing and detoxifying enzymes of carbonyl compounds are discussed. The mechanistic roles of carbonyl compounds and advanced glycation end products (AGEs) in atherosclerosis, insulin resistance, thrombogenicity, and endothelial dysfunction in animal and cell culture model of DM and patients with DM are summarised. Because of the essential role of carbonyl stress, therapeutics are aimed at scavenging, metabolizing, detoxifying, and inhibiting carbonyl compounds or AGEs so that their harmful effects are minimized. Clinically used drugs, plants extracts and miscellaneous chemical with antiglycation properties are used in an animal model of DM to alleviates the impact of carbonyl compounds. Extensive clinical trials with derivatisation of available antiglycation agents to increase the bioavailability and decrease side effects are warranted further.
Collapse
Affiliation(s)
- Sushmita Bora
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605 006, India
| | - Prashant Shankarrao Adole
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605 006, India.
| |
Collapse
|
27
|
Francisco FA, Saavedra LPJ, Junior MDF, Barra C, Matafome P, Mathias PCF, Gomes RM. Early AGEing and metabolic diseases: is perinatal exposure to glycotoxins programming for adult-life metabolic syndrome? Nutr Rev 2021; 79:13-24. [PMID: 32951053 DOI: 10.1093/nutrit/nuaa074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Perinatal early nutritional disorders are critical for the developmental origins of health and disease. Glycotoxins, or advanced glycation end-products, and their precursors such as the methylglyoxal, which are formed endogenously and commonly found in processed foods and infant formulas, may be associated with acute and long-term metabolic disorders. Besides general aspects of glycotoxins, such as their endogenous production, exogenous sources, and their role in the development of metabolic syndrome, we discuss in this review the sources of perinatal exposure to glycotoxins and their involvement in metabolic programming mechanisms. The role of perinatal glycotoxin exposure in the onset of insulin resistance, central nervous system development, cardiovascular diseases, and early aging also are discussed, as are possible interventions that may prevent or reduce such effects.
Collapse
Affiliation(s)
- Flávio A Francisco
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, PR, Brazil
| | - Lucas P J Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, PR, Brazil
| | - Marcos D F Junior
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Cátia Barra
- Institute of Physiology and Coimbra Institute of Clinical and Biomedical Research, Faculty of Medicine, and the Center for Innovative Biotechnology and Biomedicine, University of Coimbra; and the Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Institute of Physiology and Coimbra Institute of Clinical and Biomedical Research, Faculty of Medicine, and the Center for Innovative Biotechnology and Biomedicine, University of Coimbra; and the Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Paulo C F Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, PR, Brazil
| | - Rodrigo M Gomes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| |
Collapse
|
28
|
Fang X, Liu L, Zhou S, Zhu M, Wang B. N‑acetylcysteine inhibits atherosclerosis by correcting glutathione‑dependent methylglyoxal elimination and dicarbonyl/oxidative stress in the aorta of diabetic mice. Mol Med Rep 2021; 23:201. [PMID: 33495825 PMCID: PMC7821347 DOI: 10.3892/mmr.2021.11840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/08/2020] [Indexed: 12/25/2022] Open
Abstract
In diabetic animal models, high plasma/tissue levels of methylglyoxal (MG) are implicated in atherosclerosis. N-acetylcysteine (NAC) is a cysteine prodrug that replenishes intracellular glutathione (GSH) levels, which can increase the elimination of MG in diabetes mellitus (DM). The present study investigated the anti-atherosclerotic role of NAC in DM and aimed to determine whether the mechanism involved GSH-dependent MG elimination in the aorta. Apolipoprotein-E knockdown (ApoE−/−) mice injected with streptozotocin for 5 days exhibited enhanced atherosclerotic plaque size in the aortic root; notably, a high-lipid diet aggravated this alteration. NAC treatment in the drinking water for 12 weeks decreased the size of the atherosclerotic lesion, which was associated with a reduction in MG-dicarbonyl stress and oxidative stress, as indicated by decreased serum malondialdehyde levels, and increased superoxide dismutase-1 and glutathione peroxidase-1 levels in the diabetic aorta. Endothelial damage was also corrected by NAC, as indicated by an increase in the expression levels of phosphorylated (p-)Akt and p-endothelial nitric oxide synthase (eNOS) in the aorta, as well as nitric oxide (NO) in the serum. In addition, MG-treated human umbilical vein endothelial cells (HUVECs) exhibited increased reactive oxygen species and decreased antioxidant enzyme expression levels. NAC treatment corrected the alteration in HUVECs induced by MG, whereas the protective role of NAC was blocked via inhibition of GSH. These findings indicated that the diabetic aorta was more susceptible to atherosclerotic lesions compared with non-diabetic ApoE−/− mice. Furthermore, NAC may offer protection against atherosclerotic development in DM by altering aortic and systemic responses via correcting GSH-dependent MG elimination, leading to decreased oxidative stress and restoration of the p-Akt/p-eNOS pathway in the aorta.
Collapse
Affiliation(s)
- Xin Fang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lihua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shaoqiong Zhou
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Mengen Zhu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Bin Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
29
|
Xu H, Liu Q, Song X, Wang C, Wang X, Ma S, Wang X, Feng Y, Meng X, Liu X, Wang W, Lou K. Fluorophore-Promoted Facile Deprotonation and Exocyclic Five-Membered Ring Cyclization for Selective and Dynamic Tracking of Labile Glyoxals. Anal Chem 2020; 92:13829-13838. [DOI: 10.1021/acs.analchem.0c02447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Huan Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Qianqian Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiaodong Song
- Medical Laboratory Department, Hua Shan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Chao Wang
- Fluorescence Research Group, Singapore University of Technology and Design, 8 Somapah Road 487372 Singapore
| | - Xinru Wang
- School of Chemistry and Chemical Engineering, Anhui University, 111 Jiulong Road, Hefei, Anhui 230601, China
| | - Shengnan Ma
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiaolei Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yan Feng
- School of Chemistry and Chemical Engineering, Anhui University, 111 Jiulong Road, Hefei, Anhui 230601, China
| | - Xiangming Meng
- School of Chemistry and Chemical Engineering, Anhui University, 111 Jiulong Road, Hefei, Anhui 230601, China
| | - Xiaogang Liu
- Fluorescence Research Group, Singapore University of Technology and Design, 8 Somapah Road 487372 Singapore
| | - Wei Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
- Department of Pharmacology and Toxicology and BIO5 Institute, University of Arizona, Tucson, Arizona 85721-0207, United States
| | - Kaiyan Lou
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
30
|
Quantification of dicarbonyl compounds in commonly consumed foods and drinks; presentation of a food composition database for dicarbonyls. Food Chem 2020; 339:128063. [PMID: 33152865 DOI: 10.1016/j.foodchem.2020.128063] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 01/14/2023]
Abstract
Dicarbonyls are reactive precursors of advanced glycation endproducts. They are formed endogenously and during food processing. Currently, a comprehensive database on dicarbonyls in foods that covers the entire range of food groups is lacking, limiting knowledge about the amount of dicarbonyls that is ingested via food. The aim of this study was to analyze the dicarbonyls methylglyoxal (MGO), glyoxal (GO), and 3-deoxyglucosone (3-DG) in commonly-consumed products in a Western diet. We validated a UHPLC-MS/MS method to quantify MGO, GO, and 3-DG. We present a dietary dicarbonyl database of 223 foods and drinks. Total dicarbonyl concentrations were highest in dried fruit, Dutch spiced cake, and candy bars (>400 mg/kg). Total dicarbonyl concentrations were lowest in tea, dairy, light soft drinks, and rice (<10 mg/kg). The presented database of MGO, GO, and 3-DG opens the possibility to accurately estimate dietary exposure to these dicarbonyls, and explore their physiological impact on human health.
Collapse
|
31
|
Hüttl M, Markova I, Miklankova D, Makovicky P, Pelikanova T, Šeda O, Šedová L, Malinska H. Adverse Effects of Methylglyoxal on Transcriptome and Metabolic Changes in Visceral Adipose Tissue in a Prediabetic Rat Model. Antioxidants (Basel) 2020; 9:antiox9090803. [PMID: 32878255 PMCID: PMC7555565 DOI: 10.3390/antiox9090803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
Excessive methylglyoxal (MG) production contributes to metabolic and vascular changes by increasing inflammatory processes, disturbing regulatory mechanisms and exacerbating tissue dysfunction. MG accumulation in adipocytes leads to structural and functional changes. We used transcriptome analysis to investigate the effect of MG on metabolic changes in the visceral adipose tissue of hereditary hypetriglyceridaemic rats, a non-obese model of metabolic syndrome. Compared to controls, 4-week intragastric MG administration impaired glucose tolerance (p < 0.05) and increased glycaemia (p < 0.01) and serum levels of MCP-1 and TNFα (p < 0.05), but had no effect on serum adiponectin or leptin. Adipose tissue insulin sensitivity and lipolysis were impaired (p < 0.05) in MG-treated rats. In addition, MG reduced the expression of transcription factor Nrf2 (p < 0.01), which controls antioxidant and lipogenic genes. Increased expression of Mcp-1 and TNFα (p < 0.05) together with activation of the SAPK/JNK signaling pathway can promote chronic inflammation in adipose tissue. Transcriptome network analysis revealed the over-representation of genes involved in insulin signaling (Irs1, Igf2, Ide), lipid metabolism (Nr1d1, Lpin1, Lrpap1) and angiogenesis (Dusp10, Tp53inp1).
Collapse
Affiliation(s)
- Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
| | - Irena Markova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
| | - Denisa Miklankova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
| | - Pavol Makovicky
- Faculty of Education, Department of Biology, J. Selye University, 94501 Komarno, Slovakia;
| | - Terezie Pelikanova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic;
| | - Ondrej Šeda
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague, Czech Republic; (O.Š.); (L.Š.)
| | - Lucie Šedová
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague, Czech Republic; (O.Š.); (L.Š.)
| | - Hana Malinska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
- Correspondence: ; Tel.: +420-261-365-369; Fax: +420-261-363-027
| |
Collapse
|
32
|
Sourris KC, Watson A, Jandeleit-Dahm K. Inhibitors of Advanced Glycation End Product (AGE) Formation and Accumulation. Handb Exp Pharmacol 2020; 264:395-423. [PMID: 32809100 DOI: 10.1007/164_2020_391] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A range of chemically different compounds are known to inhibit the formation and accumulation of advanced glycation end products (AGEs) or disrupt associated signalling pathways. There is evidence that some of these agents can provide end-organ protection in chronic diseases including diabetes. Whilst this group of therapeutics are structurally and functionally different and have a range of mechanisms of action, they ultimately reduce the deleterious actions and the tissue burden of advanced glycation end products. To date it remains unclear if this is due to the reduction in tissue AGE levels per se or the modulation of downstream signal pathways. Some of these agents either stimulate antioxidant defence or reduce the formation of reactive oxygen species (ROS), modify lipid profiles and inhibit inflammation. A number of existing treatments for glucose lowering, hypertension and hyperlipidaemia are also known to reduce AGE formation as a by-product of their action. Targeted AGE formation inhibitors or AGE cross-link breakers have been developed and have shown beneficial effects in animal models of diabetic complications as well as other chronic conditions. However, only a few of these agents have progressed to clinical development. The failure of clinical translation highlights the importance of further investigation of the advanced glycation pathway, the diverse actions of agents which interfere with AGE formation, cross-linking or AGE receptor activation and their effect on the development and progression of chronic diseases including diabetic complications. Advanced glycation end products (AGEs) are (1) proteins or lipids that become glycated as a result of exposure to sugars or (2) non-proteinaceous oxidised lipids. They are implicated in ageing and the development, or worsening, of many degenerative diseases, such as diabetes, atherosclerosis, chronic kidney and Alzheimer's disease. Several antihypertensive and antidiabetic agents and statins also indirectly lower AGEs. Direct AGE inhibitors currently investigated include pyridoxamine and epalrestat, the inhibition of the formation of reactive dicarbonyls such as methylglyoxal as an important precursor of AGEs via increased activation of the detoxifying enzyme Glo-1 and inhibitors of NOX-derived ROS to reduce the AGE/RAGE signalling.
Collapse
Affiliation(s)
- Karly C Sourris
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anna Watson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Karin Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
33
|
Abstract
Diabetes mellitus predisposes affected individuals to a significant spectrum of cardiovascular complications, one of the most debilitating in terms of prognosis is heart failure. Indeed, the increasing global prevalence of diabetes mellitus and an aging population has given rise to an epidemic of diabetes mellitus-induced heart failure. Despite the significant research attention this phenomenon, termed diabetic cardiomyopathy, has received over several decades, understanding of the full spectrum of potential contributing mechanisms, and their relative contribution to this heart failure phenotype in the specific context of diabetes mellitus, has not yet been fully resolved. Key recent preclinical discoveries that comprise the current state-of-the-art understanding of the basic mechanisms of the complex phenotype, that is, the diabetic heart, form the basis of this review. Abnormalities in each of cardiac metabolism, physiological and pathophysiological signaling, and the mitochondrial compartment, in addition to oxidative stress, inflammation, myocardial cell death pathways, and neurohumoral mechanisms, are addressed. Further, the interactions between each of these contributing mechanisms and how they align to the functional, morphological, and structural impairments that characterize the diabetic heart are considered in light of the clinical context: from the disease burden, its current management in the clinic, and where the knowledge gaps remain. The need for continued interrogation of these mechanisms (both known and those yet to be identified) is essential to not only decipher the how and why of diabetes mellitus-induced heart failure but also to facilitate improved inroads into the clinical management of this pervasive clinical challenge.
Collapse
Affiliation(s)
- Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), Parkville, Victoria 3052, Australia
| | - E. Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States
| |
Collapse
|
34
|
Zunkel K, Simm A, Bartling B. Long-term intake of the reactive metabolite methylglyoxal is not toxic in mice. Food Chem Toxicol 2020; 141:111333. [PMID: 32298726 DOI: 10.1016/j.fct.2020.111333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/13/2020] [Accepted: 04/08/2020] [Indexed: 01/09/2023]
Abstract
Reactive carbonyls, including methylglyoxal (MG), are considered toxic compounds in foodstuffs because they irreversibly modify proteins and produce advanced glycation end products (AGEs). Therefore, we studied the long-term effect of increased MG intake in mature adult mice. Six-month-old C57BL/6N mice received MG by drinking water (2.5 mg/ml; i.e., 200-300 mg/kg BW/d) until death. This treatment caused an immediate strong increase in urine MG and a delayed moderate increase in plasma MG. At 24 months of age, mice administered MG showed no changes in the blood and tissue activity of glyoxalase-1 (Glo1), an intracellular MG-detoxifying enzyme; no signs of renal insufficiency and diabetes, including unchanged AGE modifications of plasma and vessel proteins; reduced tumour incidence; and slightly increased survival. Mice simultaneously deficient in the receptor for AGEs (RAGE) and overexpressing Glo1 exhibited higher basal plasma MG levels and did generally not respond to long-term MG intake. In vitro experiments supported the minor relevance of Glo1 in the detoxification of circulating MG but the important role of plasma albumin as an MG scavenger. In conclusion, the detoxification of dietary MG through renal excretion and further mechanisms largely prevents the toxicity of MG and possibly other food-derived reactive carbonyls in mature adults.
Collapse
Affiliation(s)
- Katja Zunkel
- Department of Cardiac Surgery, Mid-German Heart Centre, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andreas Simm
- Department of Cardiac Surgery, Mid-German Heart Centre, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Babett Bartling
- Department of Cardiac Surgery, Mid-German Heart Centre, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany; Department of Animal Health Management, Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
35
|
Hanssen NMJ, Scheijen JLJM, Houben AJHM, van de Waarenburg M, Berendschot TTJM, Webers CAB, Reesink KD, van Greevenbroek MMJ, van der Kallen C, Schaper NC, Schram MT, Henry RMA, Stehouwer CDA, Schalkwijk CG. Fasting and post-oral-glucose-load levels of methylglyoxal are associated with microvascular, but not macrovascular, disease in individuals with and without (pre)diabetes: The Maastricht Study. DIABETES & METABOLISM 2020; 47:101148. [PMID: 32058030 DOI: 10.1016/j.diabet.2020.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/25/2020] [Accepted: 02/02/2020] [Indexed: 01/11/2023]
Abstract
AIMS Reactive dicarbonyl compounds, such as methylglyoxal (MGO), rise during an oral glucose tolerance test (OGTT), particularly in (pre)diabetes. Fasting MGO levels are associated with chronic kidney disease (CKD) and cardiovascular disease (CVD) in patients with poorly controlled type 2 diabetes mellitus (T2DM). Yet, whether fasting or post-OGTT plasma MGO levels are associated with vascular disease in people with (pre)diabetes is unknown. METHODS Subjects with normal glucose metabolism (n=1796; age: 57.9±8.2 years; 43.3% men), prediabetes (n=478; age: 61.6±7.6 years; 54.0% men) and T2DM (n=669; age: 63.0±7.5 years; 67.0% men) from the Maastricht Study underwent OGTTs. Plasma MGO levels were measured at baseline and 2h after OGTT by mass spectrometry. Prior CVD was established via questionnaire. CKD was reflected by estimated glomerular filtration rate (eGFR) and albuminuria; retinopathy was assessed using retinal photographs. Data were analyzed using logistic regression adjusted for gender, age, smoking, systolic blood pressure, total-to-HDL cholesterol ratio, triglycerides, HbA1c, BMI and medication use. Odd ratios (ORs) were expressed per standard deviation of LN-transformed MGO. RESULTS Fasting and post-OGTT MGO levels were associated with higher ORs for albuminuria ≥30mg/24h [fasting: 1.12 (95% CI: 0.97-1.29); post-OGTT: 1.19 (1.01-1.41)], eGFR<60mL/min/1.73 m2 [fasting: 1.58 (95% CI: 1.38-1.82), post-OGTT: 1.57 (1.34-1.83)] and retinopathy [fasting: 1.59 (95% CI: 1.01-2.53), post-OGTT: 1.38 (0.77-2.48)]. No associations with prior CVD were found. CONCLUSION Fasting and post-OGTT MGO levels were associated with microvascular disease, but not prior CVD. Thus, therapeutic strategies directed at lowering MGO levels may prevent microvascular disease.
Collapse
Affiliation(s)
- N M J Hanssen
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - J L J M Scheijen
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - A J H M Houben
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - M van de Waarenburg
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - T T J M Berendschot
- Department of Ophthalmology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - C A B Webers
- Department of Ophthalmology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - K D Reesink
- School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; Department of Biomedical Engineering, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - M M J van Greevenbroek
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - C van der Kallen
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - N C Schaper
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - M T Schram
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; Heart and Vascular Centre, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - R M A Henry
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; Heart and Vascular Centre, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - C D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - C G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
36
|
Dicarbonyl Stress and S-Glutathionylation in Cerebrovascular Diseases: A Focus on Cerebral Cavernous Malformations. Antioxidants (Basel) 2020; 9:antiox9020124. [PMID: 32024152 PMCID: PMC7071005 DOI: 10.3390/antiox9020124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/25/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Dicarbonyl stress is a dysfunctional state consisting in the abnormal accumulation of reactive α-oxaldehydes leading to increased protein modification. In cells, post-translational changes can also occur through S-glutathionylation, a highly conserved oxidative post-translational modification consisting of the formation of a mixed disulfide between glutathione and a protein cysteine residue. This review recapitulates the main findings supporting a role for dicarbonyl stress and S-glutathionylation in the pathogenesis of cerebrovascular diseases, with specific emphasis on cerebral cavernous malformations (CCM), a vascular disease of proven genetic origin that may give rise to various clinical signs and symptoms at any age, including recurrent headaches, seizures, focal neurological deficits, and intracerebral hemorrhage. A possible interplay between dicarbonyl stress and S-glutathionylation in CCM is also discussed.
Collapse
|
37
|
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: 35.2] [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
|
38
|
Maasen K, van Greevenbroek MMJ, Scheijen JLJM, van der Kallen CJH, Stehouwer CDA, Schalkwijk CG. High dietary glycemic load is associated with higher concentrations of urinary advanced glycation endproducts: the Cohort on Diabetes and Atherosclerosis Maastricht (CODAM) Study. Am J Clin Nutr 2019; 110:358-366. [PMID: 31240298 DOI: 10.1093/ajcn/nqz119] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/27/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Advanced glycation endproducts (AGEs) and their precursors (dicarbonyls) are associated with the progression of diseases such as diabetes and cardiovascular disease. Plasma concentrations of dicarbonyls methylglyoxal (MGO), glyoxal (GO), and 3-deoxyglucosone (3-DG) are increased after an oral glucose load indicating that consumption of diets high in carbohydrates may induce the endogenous formation of dicarbonyls and AGEs. OBJECTIVE To examine the associations of dietary glycemic index (GI) and glycemic load (GL) with concentrations of dicarbonyls and AGEs in plasma and urine. METHODS Cross-sectional analyses were performed in a human observational cohort [Cohort on Diabetes and Atherosclerosis Maastricht (CODAM), n = 494, 59 ± 7 y, 25% type 2 diabetes]. GI and GL were derived from FFQs. Dicarbonyls and AGEs were measured in the fasting state by ultra-performance liquid chromatography-tandem MS. MGO, GO, and 3-DG and protein-bound Nε-(carboxymethyl)lysine (CML), Nε-(1-carboxyethyl)lysine (CEL), and pentosidine were measured in plasma. Free forms of CML, CEL, and Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1) were measured in both plasma and urine. Multiple linear regression was performed with dicarbonyls and AGEs as dependent variables, and dietary GI or GL as main independent variables (all standardized). Models were adjusted for health and lifestyle factors, dietary factors, and reciprocally for GI and GL. As this was an explorative study, we did not adjust for multiple testing. RESULTS GI was not associated with any of the dicarbonyls or AGEs. GL was positively associated with free urinary MG-H1 (β = 0.34; 95% CI: 0.12, 0.55). Furthermore, GL was positively associated with free plasma MG-H1 and free urinary CML (β = 0.23; 95% CI: 0.02, 0.43; and β = 0.28; 95% CI: 0.06, 0.50), but these associations were not independent of dietary AGE intake. CONCLUSIONS A habitual diet higher in GL is associated with higher concentrations of free urinary MG-H1. This urinary AGE is most likely a reflection of AGE accumulation and degradation in tissues, where they may be involved in tissue dysfunction.
Collapse
Affiliation(s)
- Kim Maasen
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marleen M J van Greevenbroek
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jean L J M Scheijen
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Carla J H van der Kallen
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Coen D A Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Casper G Schalkwijk
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
39
|
Dicarbonyl Stress at the Crossroads of Healthy and Unhealthy Aging. Cells 2019; 8:cells8070749. [PMID: 31331077 PMCID: PMC6678343 DOI: 10.3390/cells8070749] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Dicarbonyl stress occurs when dicarbonyl metabolites (i.e., methylglyoxal, glyoxal and 3-deoxyglucosone) accumulate as a consequence of their increased production and/or decreased detoxification. This toxic condition has been associated with metabolic and age-related diseases, both of which are characterized by a pro-inflammatory and pro-oxidant state. Methylglyoxal (MGO) is the most reactive dicarbonyl and the one with the highest endogenous flux. It is the precursor of the major quantitative advanced glycated products (AGEs) in physiological systems, arginine-derived hydroimidazolones, which accumulate in aging and dysfunctional tissues. The aging process is characterized by a decline in the functional properties of cells, tissues and whole organs, starting from the perturbation of crucial cellular processes, including mitochondrial function, proteostasis and stress-scavenging systems. Increasing studies are corroborating the causal relationship between MGO-derived AGEs and age-related tissue dysfunction, unveiling a previously underestimated role of dicarbonyl stress in determining healthy or unhealthy aging. This review summarizes the latest evidence supporting a causal role of dicarbonyl stress in age-related diseases, including diabetes mellitus, cardiovascular disease and neurodegeneration.
Collapse
|
40
|
Abstract
Significance: Obesity and type 2 diabetes mellitus are increasing globally. There is also increasing associated complications, such as non-alcoholic fatty liver disease (NAFLD) and vascular complications of diabetes. There is currently no licensed treatment for NAFLD and no recent treatments for diabetic complications. New approaches are required, particularly those addressing mechanism-based risk factors for health decline and disease progression. Recent Advances: Dicarbonyl stress is the abnormal accumulation of reactive dicarbonyl metabolites such as methylglyoxal (MG) leading to cell and tissue dysfunction. It is a potential driver of obesity, diabetes, and related complications that are unaddressed by current treatments. Increased formation of MG is linked to increased glyceroneogenesis and hyperglycemia in obesity and diabetes and also down-regulation of glyoxalase 1 (Glo1)-which provides the main enzymatic detoxification of MG. Glo1 functional genomics studies suggest that increasing Glo1 expression and activity alleviates dicarbonyl stress; slows development of obesity, related insulin resistance; and prevents development of diabetic nephropathy and other microvascular complications of diabetes. A new therapeutic approach constitutes small-molecule inducers of Glo1 expression-Glo1 inducers-exploiting a regulatory antioxidant response element in the GLO1 gene. A prototype Glo1 inducer, trans-resveratrol (tRES)-hesperetin (HESP) combination, in corrected insulin resistance, improved glycemic control and vascular inflammation in healthy overweight and obese subjects in clinical trial. Critical Issues: tRES and HESP synergize pharmacologically, and HESP likely overcomes the low bioavailability of tRES by inhibition of intestinal glucuronosyltransferases. Future Directions: Glo1 inducers may now be evaluated in Phase 2 clinical trials for treatment of NAFLD and vascular complications of diabetes.
Collapse
Affiliation(s)
- Naila Rabbani
- 1 Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital , Coventry, United Kingdom .,2 Warwick Systems Biology Centre, Senate House, University of Warwick , Coventry, United Kingdom
| | - Paul J Thornalley
- 1 Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital , Coventry, United Kingdom .,2 Warwick Systems Biology Centre, Senate House, University of Warwick , Coventry, United Kingdom
| |
Collapse
|
41
|
Methylglyoxal stress, the glyoxalase system, and diabetic chronic kidney disease. Curr Opin Nephrol Hypertens 2019; 28:26-33. [DOI: 10.1097/mnh.0000000000000465] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
42
|
Yuan T, Yang T, Chen H, Fu D, Hu Y, Wang J, Yuan Q, Yu H, Xu W, Xie X. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol 2019; 20:247-260. [PMID: 30384259 PMCID: PMC6205410 DOI: 10.1016/j.redox.2018.09.025] [Citation(s) in RCA: 449] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/12/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and inflammation interact in the development of diabetic atherosclerosis. Intracellular hyperglycemia promotes production of mitochondrial reactive oxygen species (ROS), increased formation of intracellular advanced glycation end-products, activation of protein kinase C, and increased polyol pathway flux. ROS directly increase the expression of inflammatory and adhesion factors, formation of oxidized-low density lipoprotein, and insulin resistance. They activate the ubiquitin pathway, inhibit the activation of AMP-protein kinase and adiponectin, decrease endothelial nitric oxide synthase activity, all of which accelerate atherosclerosis. Changes in the composition of the gut microbiota and changes in microRNA expression that influence the regulation of target genes that occur in diabetes interact with increased ROS and inflammation to promote atherosclerosis. This review highlights the consequences of the sustained increase of ROS production and inflammation that influence the acceleration of atherosclerosis by diabetes. The potential contributions of changes in the gut microbiota and microRNA expression are discussed.
Collapse
Affiliation(s)
- Ting Yuan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Ting Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Danli Fu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yangyang Hu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Jing Wang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Qing Yuan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Hong Yu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Wenfeng Xu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
43
|
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: 7.3] [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
|
44
|
Schlotterer A, Kolibabka M, Lin J, Acunman K, Dietrich N, Sticht C, Fleming T, Nawroth P, Hammes HP. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: studies in a rat model. FASEB J 2018; 33:4141-4153. [PMID: 30485119 DOI: 10.1096/fj.201801146rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The aim of this study was to evaluate whether damage to the neurovascular unit in diabetes depends on reactive metabolites such as methylglyoxal (MG), and to assess its impact on retinal gene expression. Male Wistar rats were supplied with MG (50 mM) by drinking water and compared with age-matched streptozotocin-diabetic animals and untreated controls. Retinal damage was evaluated for the accumulation of MG-derived advanced glycation end products, changes in hexosamine and PKC pathway activation, microglial activation, vascular alterations (pericyte loss and vasoregression), neuroretinal function assessed by electroretinogram, and neurodegeneration. Retinal gene regulation was studied by microarray analysis, and transcription factor involvement was identified by upstream regulator analysis. Systemic application of MG by drinking water increased retinal MG to levels comparable with diabetic animals. Elevated retinal MG resulted in MG-derived hydroimidazolone modifications in the ganglion cell layer, inner nuclear layer, and outer nuclear layer, a moderate activation of the hexosamine pathway, a pan-retinal activation of microglia, loss of pericytes, increased formation of acellular capillaries, decreased function of bipolar cells, and increased expression of the crystallin gene family. MG mimics important aspects of diabetic retinopathy and plays a pathogenic role in microglial activation, vascular damage, and neuroretinal dysfunction. In response to MG, the retina induces expression of neuroprotective crystallins.-Schlotterer, A., Kolibabka, M., Lin, J., Acunman, K., Dietrich, N., Sticht, C., Fleming, T., Nawroth, P., Hammes, H.-P. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: studies in a rat model.
Collapse
Affiliation(s)
- Andrea Schlotterer
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Matthias Kolibabka
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jihong Lin
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Kübra Acunman
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Nadine Dietrich
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Hans-Peter Hammes
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
45
|
A causal link between oxidative stress and inflammation in cardiovascular and renal complications of diabetes. Clin Sci (Lond) 2018; 132:1811-1836. [PMID: 30166499 DOI: 10.1042/cs20171459] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Chronic renal and vascular oxidative stress in association with an enhanced inflammatory burden are determinant processes in the development and progression of diabetic complications including cardiovascular disease (CVD), atherosclerosis and diabetic kidney disease (DKD). Persistent hyperglycaemia in diabetes mellitus increases the production of reactive oxygen species (ROS) and activates mediators of inflammation as well as suppresses antioxidant defence mechanisms ultimately contributing to oxidative stress which leads to vascular and renal injury in diabetes. Furthermore, there is increasing evidence that ROS, inflammation and fibrosis promote each other and are part of a vicious connection leading to development and progression of CVD and kidney disease in diabetes.
Collapse
|
46
|
Pácal L, Chalásová K, Pleskačová A, Řehořová J, Tomandl J, Kaňková K. Deleterious Effect of Advanced CKD on Glyoxalase System Activity not Limited to Diabetes Aetiology. Int J Mol Sci 2018; 19:ijms19051517. [PMID: 29783710 PMCID: PMC5983829 DOI: 10.3390/ijms19051517] [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: 04/24/2018] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 11/17/2022] Open
Abstract
Methylglyoxal production is increased in diabetes. Methylglyoxal is efficiently detoxified by enzyme glyoxalase 1 (GLO1). The aim was to study the effect of diabetic and CKD milieu on (a) GLO1 gene expression in peripheral blood mononuclear cells; (b) GLO1 protein levels in whole blood; and (c) GLO1 activity in RBCs in vivo in diabetic vs. non-diabetic subjects with normal or slightly reduced vs. considerably reduced renal function (CKD1-2 vs. CKD3-4). A total of 83 subjects were included in the study. Gene expression was measured using real-time PCR, and protein levels were quantified using Western blotting. Erythrocyte GLO1 activity was measured spectrophotometrically. GLO1 gene expression was significantly higher in subjects with CKD1-2 compared to CKD3-4. GLO1 protein level was lower in diabetics than in non-diabetics. GLO1 activity in RBCs differed between the four groups being significantly higher in diabetics with CKD1-2 vs. healthy subjects and vs. nondiabeticsfig with CKD3-4. GLO1 activity was significantly higher in diabetics compared to nondiabetics. In conclusion, both diabetes and CKD affects the glyoxalase system. It appears that CKD in advanced stages has prevailing and suppressive effects compared to hyperglycaemia. CKD decreases GLO1 gene expression and protein levels (together with diabetes) without concomitant changes of GLO1 activity.
Collapse
Affiliation(s)
- Lukáš Pácal
- Department of Pathophysiology, Faculty of Medicine, Masaryk University Brno, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Katarína Chalásová
- Department of Pathophysiology, Faculty of Medicine, Masaryk University Brno, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Anna Pleskačová
- Department of Pathophysiology, Faculty of Medicine, Masaryk University Brno, Kamenice 5, 625 00 Brno, Czech Republic.
- Department of Biochemistry, Faculty of Medicine, Masaryk University Brno, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Jitka Řehořová
- Department of Gastroenterology, University Hospital Brno, Jihlavská 20, 625 00 Brno, Czech Republic.
| | - Josef Tomandl
- Department of Biochemistry, Faculty of Medicine, Masaryk University Brno, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Kateřina Kaňková
- Department of Pathophysiology, Faculty of Medicine, Masaryk University Brno, Kamenice 5, 625 00 Brno, Czech Republic.
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic.
| |
Collapse
|
47
|
Cooper ME, El-Osta A, Allen TJ, Watson AMD, Thomas MC, Jandeleit-Dahm KAM. Metabolic Karma-The Atherogenic Legacy of Diabetes: The 2017 Edwin Bierman Award Lecture. Diabetes 2018; 67:785-790. [PMID: 29678824 DOI: 10.2337/dbi18-0010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cardiovascular disease, despite all the recent advances in treatment of the various risk factors, remains the major cause of mortality in both type 1 and type 2 diabetes. Experimental models of diabetes-associated atherosclerosis, despite their limitations in recapitulating the human context, have assisted in the elucidation of molecular and cellular pathways implicated in the development and progression of macrovascular injury in diabetes. Our own studies have emphasized the role of oxidative stress and advanced glycation and identified potential targets for vasoprotective therapies in the setting of diabetes. Furthermore, it has been clearly shown that previous episodes of hyperglycemia play a key role in promoting end-organ injury in diabetes, as shown in clinical trials such as the UK Prospective Diabetes Study (UKPDS), Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled Evaluation Observational Study (ADVANCE-ON), and the Diabetes Control and Complications Trial/ Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC). The cause of this phenomenon, known as metabolic memory, remains to be elucidated, but it appears that epigenetic pathways, including glucose-induced histone methylation, play a central role. Further delineation of these pathways and their link to not only glucose but also other factors implicated in vascular injury should lead to more rational, potentially more effective therapies to retard diabetes-associated cardiovascular disease.
Collapse
Affiliation(s)
- Mark Emmanuel Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Assam El-Osta
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Terri Jean Allen
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | | | | | | |
Collapse
|
48
|
Reichert O, Fleming T, Neufang G, Schmelz M, Genth H, Kaever V, Wenck H, Stäb F, Terstegen L, Kolbe L, Roggenkamp D. Impaired glyoxalase activity is associated with reduced expression of neurotrophic factors and pro-inflammatory processes in diabetic skin cells. Exp Dermatol 2018; 26:44-50. [PMID: 27306297 DOI: 10.1111/exd.13118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2016] [Indexed: 12/18/2022]
Abstract
Patients suffering from type II diabetes develop several skin manifestations including cutaneous infections, diabetic dermopathy, diabetic bullae and acanthosis nigricans. Diabetic micro- and macroangiopathy as well as diabetic neuropathy are believed to play a crucial role in the development of diabetic skin disorders. A reduced cutaneous nerve fibre density was reported in diabetic subjects, which subsequently leads to impaired sensory nerve functions. Using an innervated skin model, we investigated the impact of human diabetic dermal fibroblasts and keratinocytes on porcine sensory neurons. Diabetic skin cells showed a reduced capacity to induce neurite outgrowth due to a decreased support with neurotrophic factors, such as NGF. Furthermore, diabetic keratinocytes displayed insulin resistance and increased expression of pro-inflammatory cytokines demonstrating the persistent effect of diabetes mellitus on human skin cells. Dysregulations were related to a significantly reduced glyoxalase enzyme activity in diabetic keratinocytes as experimentally reduced glyoxalase activity mimicked the increase in pro-inflammatory cytokine expression and reduction in NGF. Our results demonstrate an impaired crosstalk of diabetic skin cells and sensory neurons favouring hypo-innervation. We suggest that reduced methylglyoxal detoxification contributes to an impaired neurocutaneous interaction in diabetic skin.
Collapse
Affiliation(s)
- Olga Reichert
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Gitta Neufang
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Martin Schmelz
- Department of Anesthesiology Mannheim, Heidelberg University, Mannheim, Germany
| | - Harald Genth
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Volkhard Kaever
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Horst Wenck
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Franz Stäb
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Lara Terstegen
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Ludger Kolbe
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | | |
Collapse
|
49
|
Lin JA, Wu CH, Yen GC. Methylglyoxal displays colorectal cancer-promoting properties in the murine models of azoxymethane and CT26 isografts. Free Radic Biol Med 2018; 115:436-446. [PMID: 29269310 DOI: 10.1016/j.freeradbiomed.2017.12.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/04/2017] [Accepted: 12/15/2017] [Indexed: 12/18/2022]
Abstract
Methylglyoxal (MG), a highly reactive carbonyl species (RCS) with pro-oxidant and proinflammatory properties, may be a colon tumor-promoting factor in food and biological systems. In the present study, we found that consumption of MG significantly deteriorated azoxymethane (AOM)-induced colonic preneoplastic lesions in ICR mice, in which biomarkers of oxidative stress and inflammation within the body and feces induced by MG-fueled carbonyl stress may have played important roles. Interestingly, exposure to MG also led to increases in the serum low-density lipoprotein (LDL)/high-density lipoprotein (HDL) ratio and fecal bile acid levels in mice, which may be critical factors involved in MG-induced colonic lesions. Additionally, MG treatment (50mg/kg body weight (BW); intraperitoneally) promoted tumor growth of CT26 isografts in mice partly by carbonyl stress-evoked protumorigenic responses, including low-grade inflammation and oxidative stress. Furthermore, primary tumor cells isolated from mice with MG-induced CT26 isografts had greater proliferative and migratory activities as well as stem-like properties compared to those isolated from the vehicle controls. Excitingly, enhanced expression or activation of proteins that modulate cell survival, proliferation, or migration/invasion was also observed in those cells. In conclusion, it is conceivable that MG-induced carbonyl stress may be the pivotal promoter involved in colon cancer progression.
Collapse
Affiliation(s)
- Jer-An Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan, ROC
| | - Chi-Hao Wu
- Department of Human Development and Family Studies, National Taiwan Normal University, 162, Section 1, Heping E. Rd., Taipei City 106, Taiwan, ROC
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan, ROC; Graduate Institute of Food Safety, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan, ROC.
| |
Collapse
|
50
|
Hammes HP. Diabetic retinopathy: hyperglycaemia, oxidative stress and beyond. Diabetologia 2018; 61:29-38. [PMID: 28942458 DOI: 10.1007/s00125-017-4435-8] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 07/04/2017] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy remains a relevant clinical problem. In parallel with diagnostic and therapeutic improvements, the role of glycaemia and reactive metabolites causing cell stress and biochemical abnormalities as treatment targets needs continuous re-evaluation. Furthermore, the basic mechanisms of physiological angiogenesis, remodelling and pruning give important clues about the origins of vasoregression during the very early stages of diabetic retinopathy and can be modelled in animals. This review summarises evidence supporting a role for the neurovascular unit-composed of neuronal, glial and vascular cells-as a responder to the biochemical changes imposed by reactive metabolites and high glucose. Normoglycaemic animal models developing retinal degeneration, provide valuable information about common pathways downstream of progressive neuronal damage that induce vasoregression, as in diabetic models. These models can serve to assess novel treatments addressing the entire neurovascular unit for the benefit of early diabetic retinopathy.
Collapse
Affiliation(s)
- Hans-Peter Hammes
- 5. Med. Department, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany.
| |
Collapse
|