1
|
Zhang Y, Zhang Z, Tu C, Chen X, He R. Advanced Glycation End Products in Disease Development and Potential Interventions. Antioxidants (Basel) 2025; 14:492. [PMID: 40298887 PMCID: PMC12024296 DOI: 10.3390/antiox14040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Advanced glycation end products (AGEs) are a group of compounds formed through non-enzymatic reactions between reducing sugars and proteins, lipids, or nucleic acids. AGEs can be generated in the body or introduced through dietary sources and smoking. Recent clinical and animal studies have highlighted the significant role of AGEs in various health conditions. These compounds accumulate in nearly all mammalian tissues and are associated with a range of diseases, including diabetes and its complications, cardiovascular disease, and neurodegeneration. This review summarizes the major diseases linked to AGE accumulation, presenting both clinical and experimental evidence. The pathologies induced by AGEs share common mechanisms across different organs, primarily involving oxidative stress, chronic inflammation, and direct protein cross-linking. Interventions targeting AGE-related diseases focus on inhibiting AGE formation using synthetic or natural antioxidants, as well as reducing dietary AGE intake through lifestyle modifications. AGEs are recognized as significant risk factors that impact health and accelerate aging, particularly in individuals with hyperglycemia. Monitoring AGE level and implementing nutritional interventions can help maintain overall health and reduce the risk of AGE-related complications.
Collapse
Affiliation(s)
- Yihan Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 510006, China; (Y.Z.); (Z.Z.)
- BYHEALTH Institute of Nutrition & Health, No. 916, Huangpu Avenue East, Huangpu District, Guangzhou 510799, China; (C.T.); (X.C.)
| | - Zhen Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 510006, China; (Y.Z.); (Z.Z.)
- BYHEALTH Institute of Nutrition & Health, No. 916, Huangpu Avenue East, Huangpu District, Guangzhou 510799, China; (C.T.); (X.C.)
| | - Chuyue Tu
- BYHEALTH Institute of Nutrition & Health, No. 916, Huangpu Avenue East, Huangpu District, Guangzhou 510799, China; (C.T.); (X.C.)
| | - Xu Chen
- BYHEALTH Institute of Nutrition & Health, No. 916, Huangpu Avenue East, Huangpu District, Guangzhou 510799, China; (C.T.); (X.C.)
| | - Ruikun He
- BYHEALTH Institute of Nutrition & Health, No. 916, Huangpu Avenue East, Huangpu District, Guangzhou 510799, China; (C.T.); (X.C.)
| |
Collapse
|
2
|
Uribarri J, Calvo MS. Does the Maillard Reaction Formation of Dietary Advanced Glycation End Products With Frying Also Merit Study as a Pathogenic Mechanism of Fried Food Toxicity or Does This Toxicity Only Involve Lipid Peroxidation? J Nutr 2024; 154:2901-2903. [PMID: 39216789 DOI: 10.1016/j.tjnut.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Jaime Uribarri
- Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Mona S Calvo
- Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States.
| |
Collapse
|
3
|
Khalid M, Adem A. The dynamic roles of advanced glycation end products. VITAMINS AND HORMONES 2024; 125:1-29. [PMID: 38997161 DOI: 10.1016/bs.vh.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are a heterogeneous group of potentially harmful molecules that can form as a result of a non-enzymatic reaction between reducing sugars and proteins, lipids, or nucleic acids. The total body pool of AGEs reflects endogenously produced AGEs as well as exogeneous AGEs that come from sources such as diet and the environment. Engagement of AGEs with their cellular receptor, the receptor for advanced glycation end products (RAGE), which is expressed on the surface of various cell types, converts a brief pulse of cellular activation to sustained cellular dysfunction and tissue destruction. The AGEs/RAGE interaction triggers a cascade of intracellular signaling pathways such as mitogen-activated protein kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinases, transforming growth factor beta, c-Jun N-terminal kinases (JNK), and nuclear factor kappa B, which leads to the production of pro-inflammatory cytokines, chemokines, adhesion molecules, and oxidative stress. All these events contribute to the progression of several chronic diseases. This chapter will provide a comprehensive understanding of the dynamic roles of AGEs in health and disease which is crucial to develop interventions that prevent and mitigate the deleterious effects of AGEs accumulation.
Collapse
Affiliation(s)
- Mariyam Khalid
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Abdu Adem
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
4
|
Johansen VBI, Josefsen K, Antvorskov JC. The Impact of Dietary Factors during Pregnancy on the Development of Islet Autoimmunity and Type 1 Diabetes: A Systematic Literature Review. Nutrients 2023; 15:4333. [PMID: 37892409 PMCID: PMC10609322 DOI: 10.3390/nu15204333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
AIMS AND HYPOTHESIS The incidence of type 1 diabetes mellitus in children is considerably increasing in western countries. Thus, identification of the environmental determinants involved could ultimately lead to disease prevention. Here, we aimed to systematically review (PROSPERO ID: CRD42022362522) the current evidence of the association between maternal dietary factors during gestation and the risk of developing type 1 diabetes and/or islet autoimmunity (IA) in murine and human offspring. METHODS In accordance with PRISMA guidelines, the present systematic review searched PubMed and Scopus (n = 343) for different combinations of MeSH terms, such as type 1 diabetes, diet, islet autoimmunity, prenatal, nutrient, gluten, gliadin, vitamin, milk, and fibers. RESULTS We found that the most investigated dietary factors in the present literature were gluten, dietary advanced glycosylated end products (dAGEs), vitamin D, fatty acids, and iron. The results concerning prenatal exposure to a gluten-free environment showed a consistently protective effect on the development of IA. Prenatal exposures to vitamin D and certain fatty acids appeared to protect against the development of IA, whereas in utero iron and fat exposures correlated with increased risks of IA. CONCLUSION We conclude that a definite association is not established for most factors investigated as the literature represents a heterogeneous pool of data, although fetal exposures to some maternal dietary components, such as gluten, show consistent associations with increased risks of IA. We suggest that human prospective dietary intervention studies in both cohort and clinical settings are crucial to better evaluate critical and protective prenatal exposures from the maternal diet during pregnancy.
Collapse
Affiliation(s)
- Valdemar Brimnes Ingemann Johansen
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Department of Biology, Faculty of Science, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark; (K.J.); (J.C.A.)
| | - Knud Josefsen
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark; (K.J.); (J.C.A.)
| | - Julie Christine Antvorskov
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark; (K.J.); (J.C.A.)
- Steno Diabetes Center, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark
| |
Collapse
|
5
|
Krisanits BA, Schuster R, Randise J, Nogueira LM, Lane JT, Panguluri GA, Li H, Helke K, Cuitiño MC, Koivisto C, Spruill L, Ostrowski MC, Anderson SM, Turner DP, Findlay VJ. Pubertal exposure to dietary advanced glycation end products disrupts ductal morphogenesis and induces atypical hyperplasia in the mammary gland. Breast Cancer Res 2023; 25:118. [PMID: 37803429 PMCID: PMC10559657 DOI: 10.1186/s13058-023-01714-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Advanced glycation end products (AGEs) are reactive metabolites intrinsically linked with modern dietary patterns. Processed foods, and those high in sugar, protein and fat, often contain high levels of AGEs. Increased AGE levels are associated with increased breast cancer risk, however their significance has been largely overlooked due to a lack of direct cause-and-effect relationship. METHODS To address this knowledge gap, FVB/n mice were fed regular, low AGE, and high AGE diets from 3 weeks of age and mammary glands harvested during puberty (7 weeks) or adulthood (12 weeks and 7 months) to determine the effects upon mammary gland development. At endpoint mammary glands were harvested and assessed histologically (n ≥ 4). Immunohistochemistry and immunofluorescence were used to assess cellular proliferation and stromal fibroblast and macrophage recruitment. The Kruskal-Wallis test were used to compare continuous outcomes among groups. Mammary epithelial cell migration and invasion in response to AGE-mediated fibroblast activation was determined in two-compartment co-culture models. In vitro experiments were performed in triplicate. The nonparametric Wilcoxon rank sum test was used to compare differences between groups. RESULTS Histological analysis revealed the high AGE diet delayed ductal elongation, increased primary branching, as well as increased terminal end bud number and size. The high AGE diet also led to increased recruitment and proliferation of stromal cells to abnormal structures that persisted into adulthood. Atypical hyperplasia was observed in the high AGE fed mice. Ex vivo fibroblasts from mice fed dietary-AGEs retain an activated phenotype and promoted epithelial migration and invasion of non-transformed immortalized and tumor-derived mammary epithelial cells. Mechanistically, we found that the receptor for AGE (RAGE) is required for AGE-mediated increases in epithelial cell migration and invasion. CONCLUSIONS We observed a disruption in mammary gland development when mice were fed a diet high in AGEs. Further, both epithelial and stromal cell populations were impacted by the high AGE diet in the mammary gland. Educational, interventional, and pharmacological strategies to reduce AGEs associated with diet may be viewed as novel disease preventive and/or therapeutic initiatives during puberty.
Collapse
Affiliation(s)
- Bradley A Krisanits
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Reid Schuster
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jaime Randise
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Lourdes M Nogueira
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jackson T Lane
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Gowtami A Panguluri
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Hong Li
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Kristi Helke
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Maria C Cuitiño
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- College of Health Sciences, Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Christopher Koivisto
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Laura Spruill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David P Turner
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Victoria J Findlay
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
6
|
Yadav N, Palkhede JD, Kim SY. Anti-Glucotoxicity Effect of Phytoconstituents via Inhibiting MGO-AGEs Formation and Breaking MGO-AGEs. Int J Mol Sci 2023; 24:7672. [PMID: 37108833 PMCID: PMC10141761 DOI: 10.3390/ijms24087672] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
The therapeutic benefits of phytochemicals in the treatment of various illnesses and disorders are well documented. They show significant promise for the discovery and creation of novel medications for treating a variety of human diseases. Numerous phytoconstituents have shown antibiotic, antioxidant, and wound-healing effects in the conventional system. Traditional medicines based on alkaloids, phenolics, tannins, saponins, terpenes, steroids, flavonoids, glycosides, and phytosterols have been in use for a long time and are crucial as alternative treatments. These phytochemical elements are crucial for scavenging free radicals, capturing reactive carbonyl species, changing protein glycation sites, inactivating carbohydrate hydrolases, fighting pathological conditions, and accelerating the healing of wounds. In this review, 221 research papers have been reviewed. This research sought to provide an update on the types and methods of formation of methylglyoxal-advanced glycation end products (MGO-AGEs) and molecular pathways induced by AGEs during the progression of the chronic complications of diabetes and associated diseases as well as to discuss the role of phytoconstituents in MGO scavenging and AGEs breaking. The development and commercialization of functional foods using these natural compounds can provide potential health benefits.
Collapse
Affiliation(s)
- Neera Yadav
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea
- School of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jyoti Dnyaneshwar Palkhede
- Department of Chemistry, College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Sun-Yeou Kim
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea
| |
Collapse
|
7
|
Wächter K, Gohde B, Szabó G, Simm A. Rye Bread Crust as an Inducer of Antioxidant Genes and Suppressor of NF-κB Pathway In Vivo. Nutrients 2022; 14:nu14224790. [PMID: 36432475 PMCID: PMC9697834 DOI: 10.3390/nu14224790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Heat-processed food, like bread, containing high amounts of advanced glycation end products (AGEs), is controversially discussed regarding the effects on health and disease. In in vitro and in vivo experiments, AGEs can induce proinflammatory NF-κB and/or the anti-inflammatory NRF2 pathways. The aim of this study was to investigate how gene expression is influenced in vivo upon short as well as long-term feeding of mice with control and bread crust-food (BC). For that, the liver, kidney and heart from two days- and eight days-fed mice were isolated and gene arrays were performed. Fewer genes were affected in terms of expression after two days of BC feeding than after eight days. We observed, especially in the heart and to lesser extent in the liver, an induction of antioxidant response by BC. Among the significantly up-regulated genes identified in the heart were transcripts encoding for cardioprotective and antioxidative proteins like metallothionein 2, uncoupling protein 3 and pyruvate dehydrogenase kinase 4. In contrast, in the liver, genes encoding for inflammatory drivers like thioredoxin-interacting protein, lncRNA Mtss1 and ubiquitin-specific protease 2 were down-modulated. However, an increased expression of immunoglobulins was observed in the kidney. Furthermore, in vivo imaging analyses with NF-κB-luciferase-reporter mice uncovered a rather anti-inflammatory response, especially after three and seven days of the feeding study. Our results suggest that bread crust exerts antioxidant and anti-inflammatory effects in the model organism mouse in an organ-specific manner.
Collapse
Affiliation(s)
- Kristin Wächter
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University, Halle-Wittenberg, 06120 Halle (Saale), Germany
- Correspondence: ; Tel.: +49-345-557-7068
| | - Birte Gohde
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University, Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Gábor Szabó
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University, Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Andreas Simm
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University, Halle-Wittenberg, 06120 Halle (Saale), Germany
- Center for Medical Basic Research, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| |
Collapse
|
8
|
Du C, Whiddett RO, Buckle I, Chen C, Forbes JM, Fotheringham AK. Advanced Glycation End Products and Inflammation in Type 1 Diabetes Development. Cells 2022; 11:3503. [PMID: 36359899 PMCID: PMC9657002 DOI: 10.3390/cells11213503] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/18/2022] [Accepted: 10/31/2022] [Indexed: 08/08/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which the β-cells of the pancreas are attacked by the host's immune system, ultimately resulting in hyperglycemia. It is a complex multifactorial disease postulated to result from a combination of genetic and environmental factors. In parallel with increasing prevalence of T1D in genetically stable populations, highlighting an environmental component, consumption of advanced glycation end products (AGEs) commonly found in in Western diets has increased significantly over the past decades. AGEs can bind to cell surface receptors including the receptor for advanced glycation end products (RAGE). RAGE has proinflammatory roles including in host-pathogen defense, thereby influencing immune cell behavior and can activate and cause proliferation of immune cells such as islet infiltrating CD8+ and CD4+ T cells and suppress the activity of T regulatory cells, contributing to β-cell injury and hyperglycemia. Insights from studies of individuals at risk of T1D have demonstrated that progression to symptomatic onset and diagnosis can vary, ranging from months to years, providing a window of opportunity for prevention strategies. Interaction between AGEs and RAGE is believed to be a major environmental risk factor for T1D and targeting the AGE-RAGE axis may act as a potential therapeutic strategy for T1D prevention.
Collapse
Affiliation(s)
- Chenping Du
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| | - Rani O. Whiddett
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
| | - Irina Buckle
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| | - Josephine M. Forbes
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg 3084, Australia
| | - Amelia K. Fotheringham
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
9
|
Leung SS, Borg DJ, McCarthy DA, Boursalian TE, Cracraft J, Zhuang A, Fotheringham AK, Flemming N, Watkins T, Miles JJ, Groop PH, Scheijen JL, Schalkwijk CG, Steptoe RJ, Radford KJ, Knip M, Forbes JM. Soluble RAGE Prevents Type 1 Diabetes Expanding Functional Regulatory T Cells. Diabetes 2022; 71:1994-2008. [PMID: 35713929 PMCID: PMC9862506 DOI: 10.2337/db22-0177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/23/2022] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes is an autoimmune disease with no cure, where clinical translation of promising therapeutics has been hampered by the reproducibility crisis. Here, short-term administration of an antagonist to the receptor for advanced glycation end products (sRAGE) protected against murine diabetes at two independent research centers. Treatment with sRAGE increased regulatory T cells (Tregs) within the islets, pancreatic lymph nodes, and spleen, increasing islet insulin expression and function. Diabetes protection was abrogated by Treg depletion and shown to be dependent on antagonizing RAGE with use of knockout mice. Human Tregs treated with a RAGE ligand downregulated genes for suppression, migration, and Treg homeostasis (FOXP3, IL7R, TIGIT, JAK1, STAT3, STAT5b, CCR4). Loss of suppressive function was reversed by sRAGE, where Tregs increased proliferation and suppressed conventional T-cell division, confirming that sRAGE expands functional human Tregs. These results highlight sRAGE as an attractive treatment to prevent diabetes, showing efficacy and reproducibility at multiple research centers and in human T cells.
Collapse
Affiliation(s)
- Sherman S. Leung
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Danielle J. Borg
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
- Inflammatory Disease Biology and Therapeutics, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
| | - Domenica A. McCarthy
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
| | | | | | - Aowen Zhuang
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
| | - Amelia K. Fotheringham
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Nicole Flemming
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Thomas Watkins
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - John J. Miles
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Per-Henrik Groop
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Nephrology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Jean L. Scheijen
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Casper G. Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Raymond J. Steptoe
- Diamantina Institute, The University of Queensland and Translational Research Institute, Brisbane, Australia
| | - Kristen J. Radford
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
- Cancer Immunotherapies, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Josephine M. Forbes
- Glycation and Diabetes, Mater Research, The University of Queensland and Translational Research Institute, Brisbane, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Mater Clinical School, The University of Queensland, Brisbane, Australia
| |
Collapse
|
10
|
Advanced Glycation End Products (AGEs) and Chronic Kidney Disease: Does the Modern Diet AGE the Kidney? Nutrients 2022; 14:nu14132675. [PMID: 35807857 PMCID: PMC9268915 DOI: 10.3390/nu14132675] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/13/2022] Open
Abstract
Since the 1980s, chronic kidney disease (CKD) affecting all ages has increased by almost 25%. This increase may be partially attributable to lifestyle changes and increased global consumption of a “western” diet, which is typically energy dense, low in fruits and vegetables, and high in animal protein and ultra-processed foods. These modern food trends have led to an increase in the consumption of advanced glycation end products (AGEs) in conjunction with increased metabolic dysfunction, obesity and diabetes, which facilitates production of endogenous AGEs within the body. When in excess, AGEs can be pathological via both receptor-mediated and non-receptor-mediated pathways. The kidney, as a major site for AGE clearance, is particularly vulnerable to AGE-mediated damage and increases in circulating AGEs align with risk of CKD and all-cause mortality. Furthermore, individuals with significant loss of renal function show increased AGE burden, particularly with uraemia, and there is some evidence that AGE lowering via diet or pharmacological inhibition may be beneficial for CKD. This review discusses the pathways that drive AGE formation and regulation within the body. This includes AGE receptor interactions and pathways of AGE-mediated pathology with a focus on the contribution of diet on endogenous AGE production and dietary AGE consumption to these processes. We then analyse the contribution of AGEs to kidney disease, the evidence for dietary AGEs and endogenously produced AGEs in driving pathogenesis in diabetic and non-diabetic kidney disease and the potential for AGE targeted therapies in kidney disease.
Collapse
|
11
|
Differences in kinetics and dynamics of endogenous versus exogenous advanced glycation end products (AGEs) and their precursors. Food Chem Toxicol 2022; 164:112987. [PMID: 35398182 DOI: 10.1016/j.fct.2022.112987] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/16/2022] [Accepted: 04/01/2022] [Indexed: 12/31/2022]
Abstract
Advanced glycation end products (AGEs) and their precursors, referred to as glycation products, are a heterogenous group of compounds being associated with adverse health effects. They are formed endogenously and in exogenous sources including food. This review investigates the roles of endogenously versus exogenously formed glycation products in the potential induction of adverse health effects, focusing on differences in toxicokinetics and toxicodynamics, which appeared to differ depending on the molecular mass of the glycation product. Based on the available data, exogenous low molecular mass (LMM) glycation products seem to be bioavailable and to contribute to dicarbonyl stress and protein cross-linking resulting in formation of endogenous AGEs. Bioavailability of exogenous high molecular mass (HMM) glycation products appears limited, while these bind to the AGE receptor (RAGE), initiating adverse health effects. Together, this suggests that RAGE-binding in relevant tissues will more likely result from endogenously formed glycation products. Effects on gut microbiota induced by glycation products is proposed as a third mode of action. Overall, studies which better discriminate between LMM and HMM glycation products and between endogenous and exogenous formation are needed to further elucidate the contributions of these different types and sources of glycation products to the ultimate biological effects.
Collapse
|
12
|
Merhi Z, Du XQ, Charron MJ. Postnatal weaning to different diets leads to different reproductive phenotypes in female offspring following perinatal exposure to high levels of dietary advanced glycation end products. F&S SCIENCE 2022; 3:95-105. [PMID: 35559999 DOI: 10.1016/j.xfss.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To examine, following perinatal exposure to a diet high in advanced glycation end products (AGEs), whether the use of standard AGE-free mouse chow during the postweaning period alters metabolism and reproduction differently than exposure to a diet low in AGEs. DESIGN Experimental animal study. SETTING University-based research laboratory. ANIMAL(S) Female CD1 mice. INTERVENTION(S) Seven-week-old mice were placed on a diet either low or high in AGEs perinatally, before mating and then during pregnancy and lactation. All offspring were weaned onto an AGE-free normal chow. MAIN OUTCOME MEASURE(S) Growth curve, liver and abdominal fat weight, insulin and glucose tolerance tests, vaginal opening, estrous cyclicity, and serum levels of antimüllerian hormone, leptin, and adiponectin were assessed. Ovarian histologic examination for follicular count and gene expression was also performed. RESULT(S) Compared with the mice exposed to a diet low in AGEs, the mice exposed to a diet high in AGEs showed lower body weight in pups, lower liver weight, delayed vaginal opening, higher serum antimüllerian hormone levels, lower primordial and secondary follicle pools, and higher ovarian Fshr messenger RNA levels. CONCLUSION(S) Following weaning, perinatal AGEs can target puberty onset and folliculogenesis differently to standard mouse chow.
Collapse
Affiliation(s)
- Zaher Merhi
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, SUNY Downstate Health Sciences University, Brooklyn, New York; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York.
| | - Xiu Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York; Department of Obstetrics, Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
13
|
Grilo LF, Tocantins C, Diniz MS, Gomes RM, Oliveira PJ, Matafome P, Pereira SP. Metabolic Disease Programming: From Mitochondria to Epigenetics, Glucocorticoid Signalling and Beyond. Eur J Clin Invest 2021; 51:e13625. [PMID: 34060076 DOI: 10.1111/eci.13625] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
Embryonic and foetal development are critical periods of development in which several environmental cues determine health and disease in adulthood. Maternal conditions and an unfavourable intrauterine environment impact foetal development and may programme the offspring for increased predisposition to metabolic diseases and other chronic pathologic conditions throughout adult life. Previously, non-communicable chronic diseases were only associated with genetics and lifestyle. Now the origins of non-communicable chronic diseases are associated with early-life adaptations that produce long-term dysfunction. Early-life environment sets the long-term health and disease risk and can span through multiple generations. Recent research in developmental programming aims at identifying the molecular mechanisms responsible for developmental programming outcomes that impact cellular physiology and trigger adulthood disease. The identification of new therapeutic targets can improve offspring's health management and prevent or overcome adverse consequences of foetal programming. This review summarizes recent biomedical discoveries in the Developmental Origins of Health and Disease (DOHaD) hypothesis and highlight possible developmental programming mechanisms, including prenatal structural defects, metabolic (mitochondrial dysfunction, oxidative stress, protein modification), epigenetic and glucocorticoid signalling-related mechanisms suggesting molecular clues for the causes and consequences of programming of increased susceptibility of offspring to metabolic disease after birth. Identifying mechanisms involved in DOHaD can contribute to early interventions in pregnancy or early childhood, to re-set the metabolic homeostasis and break the chain of subsequent events that could lead to the development of disease.
Collapse
Affiliation(s)
- Luís F Grilo
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carolina Tocantins
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana S Diniz
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rodrigo Mello Gomes
- Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, Brazil
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Department of Complementary Sciences, Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Susana P Pereira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Laboratory of Metabolism and Exercise (LametEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
14
|
Experimental Animal Studies Support the Role of Dietary Advanced Glycation End Products in Health and Disease. Nutrients 2021; 13:nu13103467. [PMID: 34684468 PMCID: PMC8539226 DOI: 10.3390/nu13103467] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022] Open
Abstract
The increased incidence of obesity, diabetes mellitus, aging, and associated comorbidities indicates the interplay between genetic and environmental influences. Several dietary components have been identified to play a role in the pathogenesis of the so-called "modern diseases", and their complications including advanced glycation end products (AGEs), which are generated during the food preparation and processing. Diet-derived advanced glycation end products (dAGEs) can be absorbed in the gastrointestinal system and contribute to the total body AGEs' homeostasis, partially excreted in the urine, while a significant amount accumulates to various tissues. Various in vitro, in vivo, and clinical studies support that dAGEs play an important role in health and disease, in a similar way to those endogenously formed. Animal studies using wild type, as well as experimental, animal models have shown that dAGEs contribute significantly to the pathogenesis of various diseases and their complications, and are involved in the changes related to the aging process. In addition, they support that dAGEs' restriction reduces insulin resistance, oxidative stress, and inflammation; restores immune alterations; and prevents or delays the progression of aging, obesity, diabetes mellitus, and their complications. These data can be extrapolated in humans and strongly support that dAGEs' restriction should be considered as an alternative therapeutic intervention.
Collapse
|
15
|
Merhi Z, Du XQ, Charron MJ. Perinatal exposure to high dietary advanced glycation end products affects the reproductive system in female offspring in mice. Mol Hum Reprod 2021; 26:615-623. [PMID: 32609365 DOI: 10.1093/molehr/gaaa046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Maternal nutrition and the intrauterine environment are important in determining susceptibility to reproductive and metabolic disturbances. Advanced glycation end products (AGEs) are widely consumed in Western diet. The purpose of this study was to determine whether perinatal exposure to a high levels of dietary AGEs affect metabolic and reproductive parameters in female mice offspring. Female CD1 mice, 7 weeks old, were placed on either a diet low (L-AGE) or high (H-AGE) in AGEs before mating and then during pregnancy and lactation. All offspring were weaned onto the L-AGE diet and studied through to 16 weeks of age; they were counted and weighed at birth and then every week for a total of 11 weeks. Vaginal opening, litter size, growth curve, liver and abdominal fat weights, serum levels of anti-Mullerian hormone, leptin and adiponectin, as well as insulin and glucose tolerance tests were compared. Ovaries were harvested for follicular count and gene expression by real-time polymerase chain reaction. Compared to perinatal exposure to the L-AGE diet, perinatal exposure to the H-AGE diet caused lower body weight at birth, and adult offspring exhibited delayed growth, lower serum leptin and adiponectin levels, delayed vaginal opening, irregular oestrous cyclicity, arrested follicular development and significant alterations in the expression of genes involved in folliculogenesis (Amh and Amhr2) and steroidogenesis (Cyp19a1). These results indicate that perinatal exposure to a diet elevated in AGEs causes deficits in perinatal growth, pubertal onset, and reproductive organ development in female mice. Whether these findings translate to humans remains to be determined in future studies.
Collapse
Affiliation(s)
- Zaher Merhi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA.,Department of Obstetrics and Gynecology NYU School of Medicine, New York, NY 10016, USA
| | - Xiu Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Obstetrics, Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Medicine & the Fleischer Institute for Diabetes & Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
16
|
Borg DJ, Faridi P, Giam KL, Reeves P, Fotheringham AK, McCarthy DA, Leung S, Ward MS, Harcourt BE, Ayala R, Scheijen JL, Briskey D, Dudek NL, Schalkwijk CG, Steptoe R, Purcell AW, Forbes JM. Short Duration Alagebrium Chloride Therapy Prediabetes Does Not Inhibit Progression to Autoimmune Diabetes in an Experimental Model. Metabolites 2021; 11:426. [PMID: 34203471 PMCID: PMC8305727 DOI: 10.3390/metabo11070426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/17/2022] Open
Abstract
Mechanisms by which advanced glycation end products (AGEs) contribute to type 1 diabetes (T1D) pathogenesis are poorly understood. Since life-long pharmacotherapy with alagebrium chloride (ALT) slows progression to experimental T1D, we hypothesized that acute ALT therapy delivered prediabetes, may be effective. However, in female, non-obese diabetic (NODShiLt) mice, ALT administered prediabetes (day 50-100) did not protect against experimental T1D. ALT did not decrease circulating AGEs or their precursors. Despite this, pancreatic β-cell function was improved, and insulitis and pancreatic CD45.1+ cell infiltration was reduced. Lymphoid tissues were unaffected. ALT pre-treatment, prior to transfer of primed GC98 CD8+ T cell receptor transgenic T cells, reduced blood glucose concentrations and delayed diabetes, suggesting islet effects rather than immune modulation by ALT. Indeed, ALT did not reduce interferon-γ production by leukocytes from ovalbumin-pre-immunised NODShiLt mice and NODscid recipients given diabetogenic ALT treated NOD splenocytes were not protected against T1D. To elucidate β-cell effects, NOD-derived MIN6N8 β-cell major histocompatibility complex (MHC) Class Ia surface antigens were examined using immunopeptidomics. Overall, no major changes in the immunopeptidome were observed during the various treatments with all peptides exhibiting allele specific consensus binding motifs. As expected, longer MHC Class Ia peptides were captured bound to H-2Db than H-2Kb under all conditions. Moreover, more 10-12 mer peptides were isolated from H-2Db after AGE modified bovine serum albumin (AGE-BSA) treatment, compared with bovine serum albumin (BSA) or AGE-BSA+ALT treatment. Proteomics of MIN6N8 cells showed enrichment of processes associated with catabolism, the immune system, cell cycling and presynaptic endocytosis with AGE-BSA compared with BSA treatments. These data show that short-term ALT intervention, given prediabetes, does not arrest experimental T1D but transiently impacts β-cell function.
Collapse
Affiliation(s)
- Danielle J. Borg
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
- Pregnancy and Development, Mater Research Institute, The University of Queensland, South Brisbane, QLD 4101, Australia
| | - Pouya Faridi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Kai Lin Giam
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Peta Reeves
- Tolerance and Autoimmunity Group, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia; (P.R.); (R.S.)
| | - Amelia K. Fotheringham
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Domenica A. McCarthy
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Sherman Leung
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Micheal S. Ward
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Brooke E. Harcourt
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Rochelle Ayala
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Jean L. Scheijen
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, 6211 Maastricht, The Netherlands; (J.L.S.); (C.G.S.)
- Cardiovascular Research Institute Maastricht, 6211 Maastricht, The Netherlands
| | - David Briskey
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD 4067, Australia;
| | - Nadine L. Dudek
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Casper G. Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, 6211 Maastricht, The Netherlands; (J.L.S.); (C.G.S.)
- Cardiovascular Research Institute Maastricht, 6211 Maastricht, The Netherlands
| | - Raymond Steptoe
- Tolerance and Autoimmunity Group, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia; (P.R.); (R.S.)
| | - Anthony W. Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Josephine M. Forbes
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
- Mater Clinical School, The University of Queensland, Brisbane, QLD 4101, Australia
| |
Collapse
|
17
|
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
|
18
|
Thornton K, Merhi Z, Jindal S, Goldsammler M, Charron MJ, Buyuk E. Dietary Advanced Glycation End Products (AGEs) could alter ovarian function in mice. Mol Cell Endocrinol 2020; 510:110826. [PMID: 32339649 DOI: 10.1016/j.mce.2020.110826] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 12/17/2022]
Abstract
Nutrition is an important source of exogenous AGEs and thermally processed foods present in western-style diets contain a large amount of these pro-inflammatory AGEs. Additionally, the intake of dietary AGEs could upregulate ovarian gene expression of inflammatory macrophage markers. The objective of this study was to investigate the effect of diet rich in AGEs on estrous cyclicity and ovarian function in a mouse model. Six-week old C57BL/6 J female mice were randomly subjected to either a diet low in AGEs (L-AGE) or a diet high in AGEs (H-AGE) for a total of 13 weeks. Experiments performed included daily vaginal smears to assess estrous cyclicity, ovarian superovulation with gonadotropins to assess the number of oocytes released, whole ovarian tissue mRNA quantification by RT-PCR to quantify genes involved in folliculogenesis, steroidogenesis, and macrophage markers, and ovarian morphology for follicle count. Outcome measures included estrous cyclicity, number of oocytes following superovulation, expression of genes involved in folliculogenesis, steroidogenesis, and macrophage infiltration as well as the number of primordial, primary, secondary, antral follicles and corpora lutea. Compared to mice on L-AGE diet, mice on H-AGE spent significantly longer time in the diestrus phase, had similar number of oocytes released following ovarian superovulation, and showed significant alterations in genes involved in steroidogenesis (increase in Star mRNA expression levels) and folliculogenesis (increase in Gdf-9 and Fshr mRNA expression levels). Mouse macrophage marker F4/80 mRNA expression was upregulated in mice on H-AGE diet compared to mice on L-AGE diet. Finally, mice on H-AGE diet had significantly fewer corpora lutea in their ovaries. These results indicate that the ingestion of high amounts of dietary AGEs could disrupt folliculogenesis and steroidogenesis that might lead to abnormal estrous cyclicity. Intake of dietary AGEs could also upregulate ovarian gene expression of inflammatory macrophage markers.
Collapse
Affiliation(s)
- Kimberly Thornton
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Currently at Reproductive Medicine Associates of New York, Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, 10022, USA
| | - Zaher Merhi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Sangita Jindal
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Michelle Goldsammler
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Maureen J Charron
- Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Erkan Buyuk
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Currently at Reproductive Medicine Associates of New York, Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, 10022, USA.
| |
Collapse
|
19
|
Krishnamoorthy D, Hoy RC, Natelson DM, Torre OM, Laudier DM, Iatridis JC, Illien-Jünger S. Dietary advanced glycation end-product consumption leads to mechanical stiffening of murine intervertebral discs. Dis Model Mech 2018; 11:dmm.036012. [PMID: 30498097 PMCID: PMC6307905 DOI: 10.1242/dmm.036012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Back pain is a leading cause of disability and is strongly associated with intervertebral disc (IVD) degeneration. Reducing structural disruption and catabolism in IVD degeneration remains an important clinical challenge. Pro-oxidant and structure-modifying advanced glycation end-products (AGEs) contribute to obesity and diabetes, which are associated with increased back pain, and accumulate in tissues due to hyperglycemia or ingestion of foods processed at high heat. Collagen-rich IVDs are particularly susceptible to AGE accumulation due to their slow metabolic rates, yet it is unclear whether dietary AGEs can cross the endplates to accumulate in IVDs. A dietary mouse model was used to test the hypothesis that chronic consumption of high AGE diets results in sex-specific IVD structural disruption and functional changes. High AGE diet resulted in AGE accumulation in IVDs and increased IVD compressive stiffness, torque range and failure torque, particularly for females. These biomechanical changes were likely caused by significantly increased AGE crosslinking in the annulus fibrosus, measured by multiphoton imaging. Increased collagen damage measured with collagen hybridizing peptide did not appear to influence biomechanical properties and may be a risk factor as these animals age. The greater influence of high AGE diet on females is an important area of future investigation that may involve AGE receptors known to interact with estrogen. We conclude that high AGE diets can be a source for IVD crosslinking and collagen damage known to be important in IVD degeneration. Dietary modifications and interventions that reduce AGEs warrant further investigation and may be particularly important for diabetics, in whom AGEs accumulate more rapidly. Summary: Dietary AGEs lead to sex-specific intervertebral disc structural and functional changes and may be targeted for promoting spinal health, especially in diabetes, in which AGEs form rapidly.
Collapse
Affiliation(s)
- Divya Krishnamoorthy
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert C Hoy
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Devorah M Natelson
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Olivia M Torre
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Damien M Laudier
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James C Iatridis
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Svenja Illien-Jünger
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
20
|
Šebeková K, Brouder Šebeková K. Glycated proteins in nutrition: Friend or foe? Exp Gerontol 2018; 117:76-90. [PMID: 30458224 DOI: 10.1016/j.exger.2018.11.012] [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] [Received: 02/22/2018] [Revised: 09/20/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022]
Abstract
Advanced glycation end products (AGEs) are formed in in vivo, and accumulate in tissues and body fluids during ageing. Endogenous AGE-modified proteins show altered structure and function, and may interact with receptor for AGEs (RAGE) resulting in production of reactive oxygen species, inflammatory, atherogenic and diabetogenic responses. AGEs are also formed in thermally processed foods. Studies in rodents document that dietary AGEs are partially absorbed into circulation, and accumulate in different tissues. Knowledge on the health effects of high dietary intake of AGEs is incomplete and contradictory. In this overview we discuss the data from experimental and clinical studies, either those supporting the assumption that restriction of dietary AGEs associated with health benefits, or data suggesting that dietary intake of AGEs associates with positive health outcomes. We polemicize whether the effects of exaggerated intake or restriction of highly thermally processed foods might be straightforward interpreted as the effects of AGEs-rich vs. AGEs-restricted diets. We also underline the lack of studies, and thus a poor knowledge, on the effects of different single chemically defined AGEs administration, concurrent intake of different dietary AGEs, of load with dietary AGEs corresponding to the habitual diet in humans, and on those of dietary AGEs in vulnerable populations, such as infants and particularly elderly.
Collapse
Affiliation(s)
- Katarína Šebeková
- Institute of Molecular Biomedicine, Medical Faculty, Comenius University, Bratislava, Slovakia.
| | - Katarína Brouder Šebeková
- Intensive Care Unit, John Radcliffe Hospital, Oxford, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
21
|
Chen JH, Lin X, Bu C, Zhang X. Role of advanced glycation end products in mobility and considerations in possible dietary and nutritional intervention strategies. Nutr Metab (Lond) 2018; 15:72. [PMID: 30337945 PMCID: PMC6180645 DOI: 10.1186/s12986-018-0306-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/21/2018] [Indexed: 02/08/2023] Open
Abstract
Advanced glycation end products (AGEs), a group of compounds that are formed by non-enzymatic reactions between carbonyl groups of reducing sugars and free amino groups of proteins, lipids or nucleic acids, can be obtained exogenously from diet or formed endogenously within the body. AGEs accumulate intracellularly and extracellularly in all tissues and body fluids and can cross-link with other proteins and thus affect their normal functions. Furthermore, AGEs can interact with specific cell surface receptors and hence alter cell intracellular signaling, gene expression, the production of reactive oxygen species and the activation of several inflammatory pathways. High levels of AGEs in diet as well as in tissues and the circulation are pathogenic to a wide range of diseases. With respect to mobility, AGEs accumulate in bones, joints and skeletal muscles, playing important roles in the development of osteoporosis, osteoarthritis, and sarcopenia with aging. This report covered the related pathological mechanisms and the potential pharmaceutical and dietary intervention strategies in reducing systemic AGEs. More prospective studies are needed to determine whether elevated serum AGEs and/or skin autofluorescence predict a decline in measures of mobility. In addition, human intervention studies are required to investigate the beneficial effects of exogenous AGEs inhibitors on mobility outcomes.
Collapse
Affiliation(s)
- Jie-Hua Chen
- Science and Technology Centre, By-Health Co. Ltd, No. 3 Kehui 3rd Street, No. 99 Kexue Avenue Central, Science City, Luogang District, Guangzhou, 510000 China
| | - Xu Lin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Cuihong Bu
- Science and Technology Centre, By-Health Co. Ltd, No. 3 Kehui 3rd Street, No. 99 Kexue Avenue Central, Science City, Luogang District, Guangzhou, 510000 China
| | - Xuguang Zhang
- Science and Technology Centre, By-Health Co. Ltd, No. 3 Kehui 3rd Street, No. 99 Kexue Avenue Central, Science City, Luogang District, Guangzhou, 510000 China
| |
Collapse
|
22
|
Khan H, Khan MS, Ahmad S. The in vivo and in vitro approaches for establishing a link between advanced glycation end products and lung cancer. J Cell Biochem 2018; 119:9099-9109. [PMID: 30076739 DOI: 10.1002/jcb.27170] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 05/18/2018] [Indexed: 01/12/2023]
Abstract
Advanced glycation end products (AGEs) are directly related to third aging-associated diseases, such as cardiovascular diseases, arteriosclerosis, and neurodegeneration. Likewise, these irreversible and nonenzymatic products have been reported to be involved in the progression of malignant cancers. In general, aging-associated diseases and the initiation of cancer have been subjects of interest for several years. Few studies on the role of AGEs in cancer have been performed on cell lines. Moreover, past investigations in the field of glycation biology still lack the knowledge of in vivo and in vitro approaches for cancer cells. Accordingly, we aimed to focus on and establish a link between cancer and glycation with respect to all the possible AGEs. In our study, the levels of carboxymethyllysine (CML) increased by 50.94% in an animal model of glycation, whereas in an animal model of cancer, the contents of CML increased by 45.94% compared with their negative controls. Similarly, fluorescent AGEs were also examined and were found to be increased by 65.3% and 58.63% in the animal models of glycation and cancer, respectively, compared with the control subjects. The protein carbonyl contents were also found to be enhanced in the animal models of glycation and cancer. In our study, the levels of reactive oxygen species were also found to be significantly increased in the in vitro model of cancer cells as compared with the controls. Such an initial breakthrough indicated that AGEs were present in the serum of the animal models of cancer and glycation.
Collapse
Affiliation(s)
- Hamda Khan
- Department of Biosciences, Integral University, Lucknow, India.,IIRC-1, Laboratory of Glycation Biology and Metabolic Disorders, Integral University, Lucknow, India
| | - Mohd Sajid Khan
- Department of Biosciences, Integral University, Lucknow, India
| | - Saheem Ahmad
- Department of Biosciences, Integral University, Lucknow, India.,IIRC-1, Laboratory of Glycation Biology and Metabolic Disorders, Integral University, Lucknow, India
| |
Collapse
|
23
|
Cerychova R, Bohuslavova R, Papousek F, Sedmera D, Abaffy P, Benes V, Kolar F, Pavlinkova G. Adverse effects of Hif1a mutation and maternal diabetes on the offspring heart. Cardiovasc Diabetol 2018; 17:68. [PMID: 29753320 PMCID: PMC5948854 DOI: 10.1186/s12933-018-0713-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/05/2018] [Indexed: 12/11/2022] Open
Abstract
Background Epidemiological studies show that maternal diabetes predisposes offspring to cardiovascular and metabolic disorders. However, the precise mechanisms for the underlying penetrance and disease predisposition remain poorly understood. We examined whether hypoxia-inducible factor 1 alpha, in combination with exposure to a diabetic intrauterine environment, influences the function and molecular structure of the adult offspring heart. Methods and results In a mouse model, we demonstrated that haploinsufficient (Hif1a+/−) offspring from a diabetic pregnancy developed left ventricle dysfunction at 12 weeks of age, as manifested by decreased fractional shortening and structural remodeling of the myocardium. Transcriptional profiling by RNA-seq revealed significant transcriptome changes in the left ventricle of diabetes-exposed Hif1a+/− offspring associated with development, metabolism, apoptosis, and blood vessel physiology. In contrast, both wild type and Hif1a+/− offspring from diabetic pregnancies showed changes in immune system processes and inflammatory responses. Immunohistochemical analyses demonstrated that the combination of haploinsufficiency of Hif1a and exposure to maternal diabetes resulted in impaired macrophage infiltration, increased levels of advanced glycation end products, and changes in vascular homeostasis in the adult offspring heart. Conclusions Together our findings provide evidence that a global reduction in Hif1a gene dosage increases predisposition of the offspring exposed to maternal diabetes to cardiac dysfunction, and also underscore Hif1a as a critical factor in the fetal programming of adult cardiovascular disease. Electronic supplementary material The online version of this article (10.1186/s12933-018-0713-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Radka Cerychova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, 25250, Vestec, Czechia.,Faculty of Science, Charles University, Prague, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, 25250, Vestec, Czechia
| | | | - David Sedmera
- Institute of Physiology CAS, Prague, Czechia.,Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czechia
| | - Vladimir Benes
- EMBL Genomics Core Facility, Meyerhofstr. 1, 69117, Heidelberg, Germany
| | | | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, 25250, Vestec, Czechia.
| |
Collapse
|
24
|
Borg DJ, Yap FYT, Keshvari S, Simmons DG, Gallo LA, Fotheringham AK, Zhuang A, Slattery RM, Hasnain SZ, Coughlan MT, Kantharidis P, Forbes JM. Perinatal exposure to high dietary advanced glycation end products in transgenic NOD8.3 mice leads to pancreatic beta cell dysfunction. Islets 2018; 10:10-24. [PMID: 29157116 PMCID: PMC5796486 DOI: 10.1080/19382014.2017.1405189] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 07/17/2017] [Accepted: 11/03/2017] [Indexed: 01/11/2023] Open
Abstract
The contribution of environmental factors to pancreatic islet damage in type 1 diabetes remains poorly understood. In this study, we crossed mice susceptible to type 1 diabetes, where parental male (CD8+ T cells specific for IGRP206-214; NOD8.3) and female (NOD/ShiLt) mice were randomized to a diet either low or high in AGE content and maintained on this diet throughout pregnancy and lactation. After weaning, NOD8.3+ female offspring were identified and maintained on the same parental feeding regimen for until day 28 of life. A low AGE diet, from conception to early postnatal life, decreased circulating AGE concentrations in the female offspring when compared to a high AGE diet. Insulin, proinsulin and glucagon secretion were greater in islets isolated from offspring in the low AGE diet group, which was akin to age matched non-diabetic C57BL/6 mice. Pancreatic islet expression of Ins2 gene was also higher in offspring from the low AGE diet group. Islet expression of glucagon, AGEs and the AGE receptor RAGE, were each reduced in low AGE fed offspring. Islet immune cell infiltration was also decreased in offspring exposed to a low AGE diet. Within pancreatic lymph nodes and spleen, the proportions of CD4+ and CD8+ T cells did not differ between groups. There were no significant changes in body weight, fasting glucose or glycemic hormones. This study demonstrates that reducing exposure to dietary AGEs throughout gestation, lactation and early postnatal life may benefit pancreatic islet secretion and immune infiltration in the type 1 diabetic susceptible mouse strain, NOD8.3.
Collapse
Affiliation(s)
- Danielle J. Borg
- Glycation and Diabetes, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Inflammatory Diseases Biology and Therapeutics, Mater Research Institute- The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Felicia Y. T. Yap
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Central and Eastern Clinical School, AMREP Precinct, Monash University, Melbourne, Australia
| | - Sahar Keshvari
- Inflammatory Diseases Biology and Therapeutics, Mater Research Institute- The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - David G. Simmons
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| | - Linda A. Gallo
- Glycation and Diabetes, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| | - Amelia K. Fotheringham
- Glycation and Diabetes, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| | - Aowen Zhuang
- Glycation and Diabetes, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Robyn M. Slattery
- Department of Immunology, Central and Eastern Clinical School, AMREP Precinct, Monash University, Melbourne, Australia
| | - Sumaira Z. Hasnain
- Inflammatory Diseases Biology and Therapeutics, Mater Research Institute- The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Melinda T. Coughlan
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Diabetes Department, Central Clinical School, Monash University, Clayton, Vic, Australia
| | - Phillip Kantharidis
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Diabetes Department, Central Clinical School, Monash University, Clayton, Vic, Australia
| | - Josephine M. Forbes
- Glycation and Diabetes, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Diabetes Department, Central Clinical School, Monash University, Clayton, Vic, Australia
- Mater Clinical School, School of Medicine, The University of Queensland, St Lucia, Australia
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Australia
| |
Collapse
|
25
|
ALjahdali N, Carbonero F. Impact of Maillard reaction products on nutrition and health: Current knowledge and need to understand their fate in the human digestive system. Crit Rev Food Sci Nutr 2017; 59:474-487. [DOI: 10.1080/10408398.2017.1378865] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Nesreen ALjahdali
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, USA
| | - Franck Carbonero
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, USA
- Department of Food Science, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
26
|
Gómez-Ojeda A, Jaramillo-Ortíz S, Wrobel K, Wrobel K, Barbosa-Sabanero G, Luevano-Contreras C, de la Maza MP, Uribarri J, Del Castillo MD, Garay-Sevilla ME. Comparative evaluation of three different ELISA assays and HPLC-ESI-ITMS/MS for the analysis of N ε-carboxymethyl lysine in food samples. Food Chem 2017; 243:11-18. [PMID: 29146316 DOI: 10.1016/j.foodchem.2017.09.098] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 08/08/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Nε-carboxymethyl-lysine (CML) is measured in food, but there is a controversy concerning the most convenient yet reliable method(s) for this task. This work compares three different ELISA assays and HPLC-ESI-ITMS/MS for the analysis of CML in several food items. The four methods showed the same decreasing order of CML concentration: beef, bacon>chicken > fish>dairy products>grain products>fruits/vegetables. HPLC-ESI-ITMS/MS results highly correlated with those obtained by ELISA performed with monoclonal CML-antibody (β=0.98, p<0.0001) whereas My Bio Source® kit results were not correlated with those provided by Lamider®. Small differences of CML concentrations in food items prepared by different culinary treatment were clearly distinguished by HPLC-ESI-ITMS/MS, but could not always be detected by ELISA. This work demonstrates a reasonable relationship between CM determined by ELISA and HPLC-ESI-ITMS/MS and therefore supports the implementation of ELISA in food CML/AGEs screening.
Collapse
Affiliation(s)
- Armando Gómez-Ojeda
- Department of Medical Science, University of Guanajuato, 20 de enero 929 Col. Obregón Leon, Guanajuato CP. 37320, Mexico.
| | - Sarahi Jaramillo-Ortíz
- Department of Chemistry, University of Guanajuato, Lascurain de Retana # 6, Centro Guanajuato, Gto., Guanajuato CP. 36000, Mexico.
| | - Katarzyna Wrobel
- Department of Chemistry, University of Guanajuato, Lascurain de Retana # 6, Centro Guanajuato, Gto., Guanajuato CP. 36000, Mexico.
| | - Kazimierz Wrobel
- Department of Chemistry, University of Guanajuato, Lascurain de Retana # 6, Centro Guanajuato, Gto., Guanajuato CP. 36000, Mexico.
| | - Gloria Barbosa-Sabanero
- Department of Medical Science, University of Guanajuato, 20 de enero 929 Col. Obregón Leon, Guanajuato CP. 37320, Mexico.
| | - Claudia Luevano-Contreras
- Department of Medical Science, University of Guanajuato, 20 de enero 929 Col. Obregón Leon, Guanajuato CP. 37320, Mexico.
| | | | - Jaime Uribarri
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Ma Dolores Del Castillo
- Food Bioscience Group, Department of Food Analysis and Bioactivity, Institute of Food Science, Research (CIAL), Spanish National Research Council (CSIC), Nicolas Cabrera 9, Madrid 28049, Spain.
| | - Ma Eugenia Garay-Sevilla
- Department of Medical Science, University of Guanajuato, 20 de enero 929 Col. Obregón Leon, Guanajuato CP. 37320, Mexico.
| |
Collapse
|
27
|
Chia JSJ, McRae JL, Kukuljan S, Woodford K, Elliott RB, Swinburn B, Dwyer KM. A1 beta-casein milk protein and other environmental pre-disposing factors for type 1 diabetes. Nutr Diabetes 2017; 7:e274. [PMID: 28504710 PMCID: PMC5518798 DOI: 10.1038/nutd.2017.16] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/20/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022] Open
Abstract
Globally type 1 diabetes incidence is increasing. It is widely accepted that the pathophysiology of type 1 diabetes is influenced by environmental factors in people with specific human leukocyte antigen haplotypes. We propose that a complex interplay between dietary triggers, permissive gut factors and potentially other influencing factors underpins disease progression. We present evidence that A1 β-casein cows’ milk protein is a primary causal trigger of type 1 diabetes in individuals with genetic risk factors. Permissive gut factors (for example, aberrant mucosal immunity), intervene by impacting the gut’s environment and the mucosal barrier. Various influencing factors (for example, breastfeeding duration, exposure to other dietary triggers and vitamin D) modify the impact of triggers and permissive gut factors on disease. The power of the dominant trigger and permissive gut factors on disease is influenced by timing, magnitude and/or duration of exposure. Within this framework, removal of a dominant dietary trigger may profoundly affect type 1 diabetes incidence. We present epidemiological, animal-based, in vitro and theoretical evidence for A1 β-casein and its β-casomorphin-7 derivative as dominant causal triggers of type 1 diabetes. The effects of ordinary milk containing A1 and A2 β-casein and milk containing only the A2 β-casein warrant comparison in prospective trials.
Collapse
Affiliation(s)
- J S J Chia
- Immunology Research Centre, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - J L McRae
- Immunology Research Centre, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - S Kukuljan
- Freedom Foods Group Ltd, Sydney, New South Wales, Australia
| | - K Woodford
- Agricultural Management Group, Lincoln University, Christchurch, New Zealand
| | - R B Elliott
- Living Cell Technologies, Auckland, New Zealand
| | - B Swinburn
- School of Population Health, University of Auckland, Auckland, New Zealand
| | - K M Dwyer
- Immunology Research Centre, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.,School of Medicine, Faculty of Health, Deakin University, Geelong, Victoria, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
28
|
Potential involvement of dietary advanced glycation end products in impairment of skeletal muscle growth and muscle contractile function in mice. Br J Nutr 2017; 117:21-29. [PMID: 28093090 DOI: 10.1017/s0007114516004591] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diets enriched with advanced glycation end products (AGE) have recently been related to muscle dysfunction processes. However, it remains unclear whether long-term exposure to an AGE-enriched diet impacts physiological characteristics of skeletal muscles. Therefore, we explored the differences in skeletal muscle mass, contractile function and molecular responses between mice receiving a diet high in AGE (H-AGE) and low in AGE (L-AGE) for 16 weeks. There were no significant differences between L-AGE and H-AGE mice with regard to body weight, food intake or epididymal fat pad weight. However, extensor digitorum longus (EDL) and plantaris (PLA) muscle weights in H-AGE mice were lower compared with L-AGE mice. Higher levels of N ε -(carboxymethyl)-l-lysine, a marker for AGE, in EDL muscles of H-AGE mice were observed compared with L-AGE mice. H-AGE mice showed lower muscle strength and endurance in vivo and lower muscle force production of PLA muscle in vitro. mRNA expression levels of myogenic factors including myogenic factor 5 and myogenic differentiation in EDL muscle were lower in H-AGE mice compared with L-AGE mice. The phosphorylation status of 70-kDa ribosomal protein S6 kinase Thr389, an indicator of protein synthesis signalling, was lower in EDL muscle of H-AGE mice than that of L-AGE mice. These findings suggest that long-term exposure to an AGE-enriched diet impairs skeletal muscle growth and muscle contractile function, and that these muscle dysfunctions may be attributed to the inhibition of myogenic potential and protein synthesis.
Collapse
|
29
|
Abstract
The receptor for advanced glycation end products (RAGE) is a novel protein increasingly studied in the pathogenesis of type 1 diabetes (T1D). RAGE is expressed by several immune cell types, including T cells, antigen-presenting cells, endothelial cells, and the endocrine cells of the pancreatic islets. RAGE binds various ligands including advanced glycation end products (AGEs), high-mobility group box protein 1 (HMGB1), S100 proteins, β-amyloid, β-sheet fibrils, and lipopolysaccharide. AGEs are a particularly interesting ligand because their exogenous introduction into the body can be accelerated by the consumption of AGE-rich processed foods. This review will detail RAGE isoforms and its ligands and discuss how RAGE binding on the aforementioned cells could be linked to T1D pathogenesis.
Collapse
Affiliation(s)
- Sherman S Leung
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Josephine M Forbes
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia.
- Mater Clinical School, School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | - Danielle J Borg
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia
| |
Collapse
|
30
|
Virtanen SM. Dietary factors in the development of type 1 diabetes. Pediatr Diabetes 2016; 17 Suppl 22:49-55. [PMID: 27411437 DOI: 10.1111/pedi.12341] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/02/2015] [Accepted: 11/06/2015] [Indexed: 12/13/2022] Open
Abstract
There are several indicators concerning the putative importance of dietary factors during the fetal period lactation, infancy and childhood in the etiology of type 1 diabetes. Among foods, cow's milk consumption has been associated with an increased risk of preclinical and/or clinical type 1 diabetes and sugars with a progression from preclinical to clinical disease. Breast milk, on the other hand, may be protective. Processed foods may be related to a greater risk of type 1 diabetes because they contain higher amounts of advanced glycation end-products. Nitrites or N-nitroso compounds in processed meat products could increase the risk of this disease. Among nutrients, n-3 fatty acids, vitamins D and E, and zinc may protect from preclinical and/or clinical type 1 diabetes. The microbial composition of foods or food's other effects on gut microbiota are receiving increasing attention, also due to their putative role in the development of type 1 diabetes. Still the number of prospective studies in this research field is limited and most of the findings remain to be replicated.
Collapse
Affiliation(s)
- Suvi M Virtanen
- Unit of Nutrition, Department of Health, National Institute for Health and Welfare, Helsinki, Finland.,School of Health Sciences, University of Tampere, Tampere, Finland.,Science Center of Pirkanmaa Hospital District, Tampere, Finland.,Center for Child Health Research, University of Tampere and University Hospital of Tampere, Tampere, Finland
| |
Collapse
|
31
|
Gupta A, Uribarri J. Dietary Advanced Glycation End Products and Their Potential Role in Cardiometabolic Disease in Children. Horm Res Paediatr 2016; 85:291-300. [PMID: 27008270 PMCID: PMC4891230 DOI: 10.1159/000444053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/12/2016] [Indexed: 12/17/2022] Open
Abstract
The rising incidence of obesity and metabolic diseases such as diabetes mellitus and cardiovascular disease in adolescents and young adults is of grave concern. Recent studies favor a role of lifestyle factors over genetics in the perpetuation of inflammation, insulin resistance and oxidative stress, which are pathophysiologic processes common to the above diseases; furthermore, the importance of dietary factors in addition to calories and physical activity in these processes is being increasingly recognized. Advanced glycation end products (AGEs) belong to a category of dietary oxidants which have been implicated in the pathogenesis of inflammation, oxidative stress, insulin resistance, β-cell failure and endothelial dysfunction. This paper reviews the studies of AGEs with a focus on their role in cardiometabolic disease in children. A Medline search was performed using the key words 'childhood obesity', 'metabolic syndrome' and 'advanced glycation end products'. Articles published in English between 1975 and 2015 and their references were reviewed. While most studies were performed in adults, a few studies also demonstrated a role of AGEs in obesity and associated cardiometabolic comorbidities in the younger population. Available evidence suggests an involvement of AGEs in the pathogenesis of adiposity and β-cell failure in children. Potential areas for further research to investigate underlying mechanisms are proposed.
Collapse
Affiliation(s)
- Anshu Gupta
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jaime Uribarri
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Abstract
AbstractDietary advanced glycation end products (AGE) formed during heating of food have gained interest as potential nutritional toxins with adverse effects on inflammation and glucose metabolism. In the present study, we investigated the short-term effects of high and low molecular weight (HMW and LMW) dietary AGE on insulin sensitivity, expression of the receptor for AGE (RAGE), the AGE receptor 1 (AGER1) and TNF-α, F2-isoprostaglandins, body composition and food intake. For 2 weeks, thirty-six Sprague–Dawley rats were fed a diet containing 20 % milk powder with different proportions of this being given as heated milk powder (0, 40 or 100 %), either native (HMW) or hydrolysed (LMW). Gene expression of RAGE and AGER1 in whole blood increased in the group receiving a high AGE LMW diet, which also had the highest urinary excretion of the AGE, methylglyoxal-derived hydroimidazolone 1 (MG-H1). Urinary excretion of Nε-carboxymethyl-lysine increased with increasing proportion of heat-treated milk powder in the HMW and LMW diets but was unrelated to gene expression. There was no difference in insulin sensitivity, F2-isoprostaglandins, food intake, water intake, body weight or body composition between the groups. In conclusion, RAGE and AGER1 expression can be influenced by a high AGE diet after only 2 weeks in proportion to MG-H1 excretion. No other short-term effects were observed.
Collapse
|
33
|
Yamagishi SI, Matsui T. Pathologic role of dietary advanced glycation end products in cardiometabolic disorders, and therapeutic intervention. Nutrition 2015; 32:157-65. [PMID: 26602289 DOI: 10.1016/j.nut.2015.08.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/03/2015] [Indexed: 01/12/2023]
Abstract
Reactive derivatives from nonenzymatic glucose-protein condensation reactions, as well as lipids and nucleic acids exposed to reducing sugars, form a heterogeneous group of irreversible adducts called AGEs (advanced glycation end products). The glycation process begins with the conversion of reversible Schiff base adducts to more stable, covalently bound Amadori rearrangement products. Over the course of days to weeks, these Amadori products undergo further rearrangement and condensation reactions to form irreversibly cross-linked macroprotein derivatives known as AGEs. The formation and accumulation of AGEs have been known to progress in a physiological aging process and at an accelerated rate under hyperglycemic and oxidative stress conditions. There is growing evidence that AGEs play a pathologic role in numerous disorders. Indeed, glycation and/or cross-linking modification of circulating or organic matrix proteins by AGEs the senescence of moieties and deteriorate their physiological function and structural integrity in multiple organ systems. Moreover, AGEs elicit oxidative stress and inflammatory reactions through the interaction with the receptor for advanced glycation products in a variety of cells, thereby contributing to the development and progression of various aging- or diabetes-related disorders, such as cardiovascular disease, chronic kidney disease, insulin resistance, and Alzheimer's disease. Recently, diet has been recognized as a major environmental source of AGEs that could cause proinflammatory reactions and organ damage in vivo. Therefore, this review summarizes the pathophysiological role of dietary AGEs in health and disease, especially focusing on cardiometabolic disorders. We also discuss the potential utility in targeting exogenously derived AGEs for therapeutic intervention.
Collapse
Affiliation(s)
- Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan.
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
34
|
Sharma C, Kaur A, Thind SS, Singh B, Raina S. Advanced glycation End-products (AGEs): an emerging concern for processed food industries. Journal of Food Science and Technology 2015; 52:7561-76. [PMID: 26604334 DOI: 10.1007/s13197-015-1851-y] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 04/12/2015] [Accepted: 04/22/2015] [Indexed: 01/02/2023]
Abstract
The global food industry is expected to increase more than US $ 7 trillion by 2014. This rise in processed food sector shows that more and more people are diverging towards modern processed foods. As modern diets are largely heat processed, they are more prone to contain high levels of advanced glycation end products (AGEs). AGEs are a group of complex and heterogeneous compounds which are known as brown and fluorescent cross-linking substances such as pentosidine, non-fluorescent cross-linking products such as methylglyoxal-lysine dimers (MOLD), or non-fluorescent, non-cross linking adducts such as carboxymethyllysine (CML) and pyrraline (a pyrrole aldehyde). The chemistry of the AGEs formation, absorption and bioavailability and their patho-biochemistry particularly in relation to different complications like diabetes and ageing discussed. The concept of AGEs receptor - RAGE is mentioned. AGEs contribute to a variety of microvascular and macrovascular complications through the formation of cross-links between molecules in the basement membrane of the extracellular matrix and by engaging the receptor for advanced glycation end products (RAGE). Different methods of detection and quantification along with types of agents used for the treatment of AGEs are reviewed. Generally, ELISA or LC-MS methods are used for analysis of foods and body fluids, however lack of universally established method highlighted. The inhibitory effect of bioactive components on AGEs by trapping variety of chemical moieties discussed. The emerging evidence about the adverse effects of AGEs makes it necessary to investigate the different therapies to inhibit AGEs.
Collapse
Affiliation(s)
- Chetan Sharma
- Department of Food Science and Technology, Punjab Agricultural University, Ludhiana, India
| | - Amarjeet Kaur
- Department of Food Science and Technology, Punjab Agricultural University, Ludhiana, India
| | - S S Thind
- Department of Food Science and Technology, Punjab Agricultural University, Ludhiana, India
| | - Baljit Singh
- Department of Food Science and Technology, Punjab Agricultural University, Ludhiana, India
| | - Shiveta Raina
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
| |
Collapse
|
35
|
Ottum MS, Mistry AM. Advanced glycation end-products: modifiable environmental factors profoundly mediate insulin resistance. J Clin Biochem Nutr 2015; 57:1-12. [PMID: 26236094 PMCID: PMC4512899 DOI: 10.3164/jcbn.15-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/13/2015] [Indexed: 12/25/2022] Open
Abstract
Advanced glycation end-products are toxic by-products of metabolism and are also acquired from high-temperature processed foods. They promote oxidative damage to proteins, lipids and nucleotides. Aging and chronic diseases are strongly associated with markers for oxidative stress, especially advanced glycation end-products, and resistance to peripheral insulin-mediated glucose uptake. Modifiable environmental factors including high levels of refined and simple carbohydrate diets, hypercaloric diets and sedentary lifestyles drive endogenous formation of advanced glycation end-products via accumulation of highly reactive glycolysis intermediates and activation of the polyol/aldose reductase pathway producing high intracellular fructose. High advanced glycation end-products overwhelm innate defenses of enzymes and receptor-mediated endocytosis and promote cell damage via the pro-inflammatory and pro-oxidant receptor for advanced glycation end-products. Oxidative stress disturbs cell signal transduction, especially insulin-mediated metabolic responses. Here we review emerging evidence that restriction of dietary advanced glycation end-products significantly reduces total systemic load and insulin resistance in animals and humans in diabetes, polycystic ovary syndrome, healthy populations and dementia. Of clinical importance, this insulin sensitizing effect is independent of physical activity, caloric intake and adiposity level.
Collapse
Affiliation(s)
- Mona S Ottum
- Dietetics and Human Nutrition Program, 318 Marshall Building, Eastern Michigan University, Ypsilanti, MI 48197, USA
| | - Anahita M Mistry
- Dietetics and Human Nutrition Program, 318 Marshall Building, Eastern Michigan University, Ypsilanti, MI 48197, USA
| |
Collapse
|
36
|
Role of nutritional factors at the early life stages in the pathogenesis and clinical course of type 1 diabetes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:382165. [PMID: 25883958 PMCID: PMC4391527 DOI: 10.1155/2015/382165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 11/02/2014] [Accepted: 11/03/2014] [Indexed: 02/06/2023]
Abstract
Nutrition has been suggested as an important environmental factor other than viruses and chemicals in the pathogenesis of type 1 diabetes (T1D). Whereas various maternal dietary nutritional elements have been suggested and examined in T1D of both humans and experimental animals, the results largely remain controversial. In a series of studies using T1D model nonobese diabetic (NOD) mice, maternal dietary n-6/n-3 essential fatty acid ratio during pregnancy and lactation period, that is, early life stages of the offspring, has been shown to affect pathogenesis of insulitis and strongly prevent overt T1D of the offspring, which is consistent with its preventive effects on other allergic diseases.
Collapse
|
37
|
Biswas SK, Mohtarin S, Mudi SR, Anwar T, Banu LA, Alam SMK, Fariduddin M, Arslan MI. Relationship of Soluble RAGE with Insulin Resistance and Beta Cell Function during Development of Type 2 Diabetes Mellitus. J Diabetes Res 2015; 2015:150325. [PMID: 26078977 PMCID: PMC4452360 DOI: 10.1155/2015/150325] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/12/2015] [Indexed: 12/25/2022] Open
Abstract
This study examined whether circulating levels of soluble receptor for advanced glycation end products (sRAGE) alter in prediabetes and correlate with insulin resistance (IR) and beta cell function in prediabetes and newly diagnosed type 2 diabetes mellitus (T2DM). Subjects without previous history of diabetes were recruited and grouped as control, prediabetes, and newly diagnosed T2DM. The control subjects (n = 40) and people with prediabetes (n = 52) and diabetes (n = 66) were similar in terms of age, sex, BMI, systolic and diastolic BP, and fasting insulin level. HOMA-IR was found significantly higher in people with diabetes than control subjects (p < 0.001) and people with prediabetes (p = 0.005); and HOMA-%B was found significantly deteriorated in people with diabetes (p < 0.001) compared to control subjects and people with prediabetes. However, serum sRAGE levels did not show any significant alteration in people with prediabetes compared to control subjects. Moreover, univariate and multivariate analyses did not identify any significant correlation and statistical association of sRAGE with HOMA-IR and HOMA-%B in people with prediabetes and newly diagnosed T2DM. Our data suggest that serum sRAGE levels do not alter in people with prediabetes compared to control subjects and do not correlate or associate with IR and beta cell function during development of T2DM.
Collapse
Affiliation(s)
- Subrata Kumar Biswas
- Department of Biochemistry, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
- *Subrata Kumar Biswas:
| | - Sabreena Mohtarin
- Department of Biochemistry, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - Sonchita Rani Mudi
- Department of Biochemistry and Cell Biology, Bangladesh Institute of Research and Rehabilitation in Diabetes, Endocrine and Metabolic Disorders (BIRDEM), 122 Kazi Nazrul Islam Avenue, Shahbag, Dhaka 1000, Bangladesh
| | - Taznuva Anwar
- Department of Biochemistry, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - Laila Anjuman Banu
- Department of Anatomy, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - Sheikh Md. Khorshed Alam
- Department of Biochemistry, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - Md. Fariduddin
- Department of Endocrinology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - M. Iqbal Arslan
- Department of Biochemistry, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| |
Collapse
|
38
|
Salonen KM, Ryhänen SJ, Forbes JM, Härkönen T, Ilonen J, Laine AP, Groop PH, Knip M. Circulating concentrations of soluble receptor for AGE are associated with age and AGER gene polymorphisms in children with newly diagnosed type 1 diabetes. Diabetes Care 2014; 37:1975-81. [PMID: 24742661 DOI: 10.2337/dc13-3049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We analyzed the relationship among soluble receptor for advanced glycation end products (sRAGEs), the clinical phenotype, HLA genotype, and risk-associated single nucleotide polymorphisms (SNPs) in the AGER gene in a large population of Finnish children with newly diagnosed type 1 diabetes. RESEARCH DESIGN AND METHODS Samples from 2,115 clinically phenotyped children <15 years of age in whom type 1 diabetes was diagnosed and 316 control subjects were analyzed for sRAGEs. Three SNPs of AGER, previously associated with HLA-DR/DQ haplotype independent diabetes risk (rs2070600, rs9469089, and rs17493811), were analyzed in 1,390 affected subjects. RESULTS Children with type 1 diabetes and control subjects had similar sRAGE concentrations (1,171 vs. 1,153 pg/mL, P = 0.48). There was a correlation between age at diagnosis and serum sRAGE concentrations (r = 0.10, P < 0.001) among the patients but not among the control subjects. Children <2 years of age had the lowest concentrations in the diabetic population (1,027 vs. 1,181 pg/mL, P < 0.001) and the highest among the control subjects (1,329 vs. 1,140 pg/mL, P = 0.04). Ketoacidosis at diagnosis was associated with reduced concentrations (1,086 vs. 1,190 pg/mL, P < 0.001). HLA DR3/DR4 heterozygosity and the DR3 allele were associated with reduced sRAGE concentrations. The predisposing AA genotype of rs2070600 was associated with decreased sRAGE concentrations, while the protective CC genotype of rs9469089 was linked to increased concentrations. CONCLUSIONS Age and AGER polymorphisms are associated with the circulating sRAGE concentration among children with type 1 diabetes. The observations of reduced sRAGE concentrations in young children, in those with ketoacidosis, and in carriers of the high-risk HLA DR3/DR4 genotype suggest that decreased sRAGE concentration reflects a more aggressive disease phenotype.
Collapse
Affiliation(s)
- Kirsi M Salonen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, FinlandDiabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
| | - Samppa J Ryhänen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, FinlandDiabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
| | - Josephine M Forbes
- Mater Research, Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Taina Härkönen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, FinlandDiabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, Turku, FinlandDepartment of Clinical Microbiology, University of Eastern Finland, Kuopio, Finland
| | | | - Per-Henrik Groop
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, FinlandFolkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, FinlandDivision of Nephrology, Department of Medicine, Helsinki University Central Hospital, Biomedicum Helsinki, Helsinki, FinlandBaker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, FinlandDiabetes and Obesity Research Program, University of Helsinki, Helsinki, FinlandFolkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, FinlandDepartment of Pediatrics, Tampere University Hospital, Tampere, Finland
| | | |
Collapse
|
39
|
Prasad C, Imrhan V, Marotta F, Juma S, Vijayagopal P. Lifestyle and Advanced Glycation End Products (AGEs) Burden: Its Relevance to Healthy Aging. Aging Dis 2014; 5:212-217. [PMID: 24900944 PMCID: PMC4037313 DOI: 10.14336/ad.2014.0500212] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 10/22/2013] [Accepted: 11/10/2013] [Indexed: 02/05/2023] Open
Abstract
Uncontrolled continued exposure to oxidative stress is a precursor to many chronic diseases including cancer, diabetes, degenerative disorders and cardiovascular diseases. Of the many known mediators of oxidative stress, reactive oxygen species (ROS) and advanced glycation end products (AGEs) are the most studied. In the present review, we have summarized current data on the origin of circulating AGEs, discussed issues associated with reliable assessment of its steady state level, and changes in its level with age and select metabolic diseases. Lastly, we have made recommendations about life style changes that may decrease AGEs burden to promote healthy aging.
Collapse
Affiliation(s)
- Chandan Prasad
- Department of Nutrition and Food Sciences, Texas Woman’s University, Denton, Texas, USA
| | - Victorine Imrhan
- Department of Nutrition and Food Sciences, Texas Woman’s University, Denton, Texas, USA
| | | | - Shanil Juma
- Department of Nutrition and Food Sciences, Texas Woman’s University, Denton, Texas, USA
| | - Parakat Vijayagopal
- Department of Nutrition and Food Sciences, Texas Woman’s University, Denton, Texas, USA
| |
Collapse
|
40
|
Abstract
Despite new and effective drug therapies, insulin resistance (IR), type 2 diabetes mellitus (T2D) and its complications remain major medical challenges. It is accepted that IR, often associated with over-nutrition and obesity, results from chronically elevated oxidant stress (OS) and chronic inflammation. Less acknowledged is that a major cause for this inflammation is excessive consumption of advanced glycation end products (AGEs) with the standard western diet. AGEs, which were largely thought as oxidative derivatives resulting from diabetic hyperglycemia, are increasingly seen as a potential risk for islet β-cell injury, peripheral IR and diabetes. Here we discuss the relationships between exogenous AGEs, chronic inflammation, IR, and T2D. We propose that under chronic exogenous oxidant AGE pressure the depletion of innate defense mechanisms is an important factor, which raises susceptibility to inflammation, IR, T2D and its complications. Finally we review evidence on dietary AGE restriction as a nonpharmacologic intervention, which effectively lowers AGEs, restores innate defenses and improves IR, thus, offering new perspectives on diabetes etiology and therapy.
Collapse
Affiliation(s)
- Helen Vlassara
- Department of Geriatrics, Mount Sinai School of Medicine, New York, NY
- Department of Medicine, Mount Sinai School of Medicine, New York, NY
| | - Jaime Uribarri
- Department of Medicine, Mount Sinai School of Medicine, New York, NY
| |
Collapse
|
41
|
Ward MS, Fotheringham AK, Cooper ME, Forbes JM. Targeting advanced glycation endproducts and mitochondrial dysfunction in cardiovascular disease. Curr Opin Pharmacol 2013; 13:654-61. [DOI: 10.1016/j.coph.2013.06.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/25/2013] [Accepted: 06/25/2013] [Indexed: 10/26/2022]
|
42
|
Poulsen MW, Hedegaard RV, Andersen JM, de Courten B, Bügel S, Nielsen J, Skibsted LH, Dragsted LO. Advanced glycation endproducts in food and their effects on health. Food Chem Toxicol 2013; 60:10-37. [PMID: 23867544 DOI: 10.1016/j.fct.2013.06.052] [Citation(s) in RCA: 543] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 06/23/2013] [Accepted: 06/26/2013] [Indexed: 12/27/2022]
Abstract
Advanced glycation endproducts (AGEs) form by Maillard-reactions after initial binding of aldehydes with amines or amides in heated foods or in living organisms. The mechanisms of formation may include ionic as well as oxidative and radical pathways. The reactions may proceed within proteins to form high-molecular weight (HMW) AGEs or among small molecules to form low-molecular weight (LMW) AGEs. All free amino acids form AGEs, but lysine or arginine side chains dominate AGE formation within proteins. The analysis of AGEs in foods and body fluids is most often performed by ELISA or LC-MS; however, none of the methodologies cover all HMW and LMW AGEs. Most research is, therefore, carried out using 'representative' AGE compounds, most often N(ε)-carboxymethyl-lysine (CML). Only LMW AGEs, including peptide-bound forms, and carbonyls may be absorbed from the gut and contribute to the body burden of AGEs. Some AGEs interact with specific pro- or anti-inflammatory receptors. Most studies on the biological effects of AGEs have been carried out by administering heated foods. The pro-inflammatory and deteriorating biological effects of AGEs in these studies, therefore, need further confirmation. The current review points out several research needs in order to address important questions on AGEs in foods and health.
Collapse
Affiliation(s)
- Malene W Poulsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Uribarri J, Cai W, Pyzik R, Goodman S, Chen X, Zhu L, Ramdas M, Striker GE, Vlassara H. Suppression of native defense mechanisms, SIRT1 and PPARγ, by dietary glycoxidants precedes disease in adult humans; relevance to lifestyle-engendered chronic diseases. Amino Acids 2013; 46:301-9. [PMID: 23636469 DOI: 10.1007/s00726-013-1502-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 04/13/2013] [Indexed: 01/12/2023]
Abstract
SIRT1 and PPARγ, host defenses regulating inflammation and metabolic functions, are suppressed under chronic high oxidant stress and inflammation (OS/Infl) conditions. In diabetes, dietary advanced glycation end products (dAGEs) cause OS/Infl and suppress SIRT1. Herein, we ask whether dAGEs also suppress host defense in adults without diabetes. The relationships between dAGEs and basal SIRT1 mRNA, PPARγ protein levels in mononuclear cells (MNC) and circulating inflammatory/metabolic markers were examined in 67 healthy adults aged >60 years and in 18 subjects, before and after random assignment to either a standard diet (regular >15 AGE Eq/day) or an isocaloric AGE-restricted diet (<10 AGE Eq/day) for 4 months. Also, the interactions of AGEs and anti-AGE receptor-1 (AGER1) with SIRT1 and PPARγ were assessed in wild type (WT) and AGER1-transduced (AGER1(+)) MNC-like THP-1 cells. We found that dAGE, but not caloric intake, correlated negatively with MNC SIRT1 mRNA levels and positively with circulating AGEs (sAGEs), OS/infl, MNC TNFα and RAGE. Basal MNC PPARγ protein was also lower in consumers of regular vs. AGE-restricted diet. AGE restriction restored MNC SIRT1 and PPARγ, and significantly decreased sAGEs, 8-isoprostanes, VCAM-1, MNC TNFα and RAGE. Model AGEs suppressed SIRT1 protein and activity, and PPARγ protein in WT, but not in AGER1(+) cells in vitro. In conclusion, chronic consumption of high-AGE diets depletes defenses such as SIRT1 and PPARγ, independent of calories, predisposing to OS/Infl and chronic metabolic disease. Restricted entry of oral AGEs may offer a disease-prevention alternative for healthy adults.
Collapse
Affiliation(s)
- Jaime Uribarri
- Department of Medicine, The Mount Sinai School of Medicine, One Gustave Levy Place, New York, NY, 10029, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Forbes JM, Cowan SP, Andrikopoulos S, Morley AL, Ward LC, Walker KZ, Cooper ME, Coughlan MT. Glucose homeostasis can be differentially modulated by varying individual components of a western diet. J Nutr Biochem 2013; 24:1251-7. [PMID: 23313044 DOI: 10.1016/j.jnutbio.2012.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 08/14/2012] [Accepted: 09/21/2012] [Indexed: 01/19/2023]
Abstract
Chronic overconsumption of a Western diet has been identified as a major risk factor for diabetes, yet precisely how each individual component contributes to defects in glucose homeostasis independent of consumption of other macronutrients remains unclear. Eight-week-old male Sprague Dawley rats were randomized to feeding with one of six semi-pure diets: control, processed (high advanced glycation end products/AGE), high protein, high dextrose (glucose polymer), high in saturated fat (plant origin), or high in saturated fat (animal origin). After chronic feeding for 24 weeks, body composition was determined by bioelectrical impedance spectroscopy and glucose homeostasis was assessed. When compared to the control and high AGE diets, excess consumption of the diet high in saturated fat (animal source) increased body weight and adiposity, and decreased insulin sensitivity, as defined by HOMA IR, impaired skeletal muscle insulin signaling and insulin hypersecretion in the context of increased circulating glucagon-like peptide (GLP-1). Compared to the control diet, chronic consumption of the high AGE, protein or dextrose diet increased fasting plasma glucose, decreased fasting plasma insulin and insulin secretion. These diets also reduced circulating GLP-1 concentrations. These data suggest that individual components of a western diet have differential effects in modulating glucose homeostasis and adiposity. These data provide clear evidence of a link between over-consumption of a western diet and the development of diabetes.
Collapse
Affiliation(s)
- Josephine M Forbes
- Diabetes Complications, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Altieri P, Cavazza C, Pasqui F, Morselli AM, Gambineri A, Pasquali R. Dietary habits and their relationship with hormones and metabolism in overweight and obese women with polycystic ovary syndrome. Clin Endocrinol (Oxf) 2013; 78:52-9. [PMID: 22288821 DOI: 10.1111/j.1365-2265.2012.04355.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 11/05/2011] [Accepted: 01/22/2012] [Indexed: 11/30/2022]
Abstract
OBJECTIVE This study investigates energy intake, macronutrient composition and habitual food choices in overweight/obese women with polycystic ovary syndrome (PCOS) and controls similar for age and body mass index (BMI), and their relationship with hormonal and metabolic parameters. DESIGN Case-control study carried out in an academic hospital in Bologna, Italy. PATIENTS One-hundred obese or overweight (BMI >25 kg/m(2) ) women with PCOS, diagnosed according to Rotterdam criteria, and 100 age- and BMI-matched controls. MEASUREMENTS Dietary habits were investigated by means of the 7 days food diary. Fasting hormones and metabolic parameters were investigated in all subjects. RESULTS We showed that diet does not differ between the two groups as regards energy, macronutrient and advanced glycosylated end product intake, except for a lower percentage of energy from lipids and a higher intake of fibres by PCOS women. PCOS women were characterized by a higher consumption of cheese and high-glycaemic index starchy sweets and a preference for raw oil rather than other cooked fats, compared to controls. The PCOS or control status influenced some of the relationships between dietary components, food choices and metabolic parameters, particularly insulin(AUC) and HDL-cholesterol. CONCLUSIONS This study did not find major differences in dietary habits between PCOS and normoandrogenic control women. Our findings support the hypothesis that specific foods may influence metabolic and hormonal pattern and that this relationship may be differently regulated in PCOS and normoandrogenic women; however, they give little support to the hypothesis of a strong dependence of PCOS status on nutritional factors.
Collapse
Affiliation(s)
- Paola Altieri
- Division of Endocrinology, S.Orsola-Malpighi Hospital, University Alma Mater Studiorum of Bologna, Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
46
|
Digestibility of extruded proteins and metabolic transit of N ε -carboxymethyllysine in rats. Amino Acids 2012; 44:1441-9. [PMID: 23160731 DOI: 10.1007/s00726-012-1427-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 11/02/2012] [Indexed: 10/27/2022]
Abstract
Milk proteins are frequently used as supplements in fortified foods. However, processing produces chemical changes which likely affect the nutritional advantage. This study was intended to explore the possible difference in digestibility between extruded and non-extruded caseins and how the dietary N (ε) -carboxymethyllysine (CML) is metabolised. Normal rats were randomized into either an extruded protein diet (EP) or the same with unextruded proteins (UEP), for two periods of 2 weeks at 7 to 9 and 11 to 13 weeks of age. However, no difference in protein digestibility was detected between the two diets, either in young or in adult animals, despite a 9.4-fold higher level of CML and an 8.5-fold higher level of lysinoalanine in the EP than in the UEP. No diet-related changes were observed in plasma CML, either protein bound or free. Amounts of 38 and 48 % of the orally absorbed CML were excreted in urine and faeces, respectively, in UEP-fed rats. Lower rates of excretion were found in the EP-fed rats (23 and 37 %, respectively). A second animal study using a single oral dose of free CML (400 μg/rat) was set up to measure the systemic concentration of CML every hour from 0 to 4 h. It revealed that protein-bound CML was not affected by the oral dose of CML, and the highest free CML level found in the circulation was 600 ng/mL. Extruded proteins, therefore, appear to be well digested, and CML rapidly eliminated. Since its elimination is, however, incomplete, the question of its biodistribution and metabolism remains open.
Collapse
|
47
|
Abstract
Type 1 diabetes (T1D) is a multi-factorial, organ-specific autoimmune disease in genetically susceptible individuals, which is characterized by a selective and progressive loss of insulin-producing β-cells. Cells mediating innate as well as adaptive immunity infiltrate pancreatic islets, thereby generating an aberrant inflammatory process called insulitis that can be mirrored by a pathologic autoantibody production and autoreactive T-cells. In tight cooperation with infiltrating innate immune cells, which secrete high levels of pro-inflammatory cytokines like IL-1β, TNFα, and INFγ effector T-cells trigger the fatal destruction process of β-cells. There is ongoing discussion on the contribution of inflammation in T1D pathogenesis, ranging from a bystander reaction of autoimmunity to a dysregulation of immune responses that initiate inflammatory processes and thereby actively promoting β-cell death. Here, we review recent advances in anti-inflammatory interventions in T1D animal models and preclinical studies and discuss their mode of action as well as their capacity to interfere with T1D development.
Collapse
Affiliation(s)
- Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Albert Einstein Allee 11, 89081, Ulm, Germany.
| | | | | |
Collapse
|
48
|
van Woudenbergh GJ, Kuijsten A, Tigcheler B, Sijbrands EJG, van Rooij FJA, Hofman A, Witteman JCM, Feskens EJM. Meat consumption and its association with C-reactive protein and incident type 2 diabetes: the Rotterdam Study. Diabetes Care 2012; 35:1499-505. [PMID: 22596177 PMCID: PMC3379589 DOI: 10.2337/dc11-1899] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate whether intake of different types of meat is associated with circulating C-reactive protein (CRP) and risk of type 2 diabetes in a prospective cohort study. RESEARCH DESIGN AND METHODS Our analysis included 4,366 Dutch participants who did not have diabetes at baseline. During a median follow-up period of 12.4 years, 456 diabetes cases were confirmed. Intake of red meat, processed meat, and poultry was derived from a food frequency questionnaire, and their association with serum high-sensitivity CRP was examined cross-sectionally using linear regression models. Their association with risk of type 2 diabetes was examined using multivariate Cox proportional hazards model, including age, sex, family history of diabetes, and lifestyle and dietary factors. RESULTS An increment of 50 g of processed meat was associated with increased CRP concentration (β(processed meat) = 0.12; P = 0.01), whereas intake of red meat and poultry was not. When comparing the highest to the lowest category of meat intake with respect to diabetes incidence, the adjusted relative risks were as follows: for red meat (1.42 [95% CI 1.06-1.91]), for processed meat (1.87 [1.26-2.78]), and for poultry (0.95 [0.74-1.22]). Additional analysis showed that the associations were not affected appreciably after inclusion of CRP into the model. After adjustment for BMI, however, the association for red meat attenuated to 1.18 (0.88-1.59). CONCLUSIONS Intake of processed meat is associated with higher risk of type 2 diabetes. It appears unlikely that CRP mediates this association.
Collapse
|
49
|
Beyan H, Riese H, Hawa MI, Beretta G, Davidson HW, Hutton JC, Burger H, Schlosser M, Snieder H, Boehm BO, Leslie RD. Glycotoxin and autoantibodies are additive environmentally determined predictors of type 1 diabetes: a twin and population study. Diabetes 2012; 61:1192-8. [PMID: 22396204 PMCID: PMC3331747 DOI: 10.2337/db11-0971] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In type 1 diabetes, diabetes-associated autoantibodies, including islet cell antibodies (ICAs), reflect adaptive immunity, while increased serum N(ε)-carboxymethyl-lysine (CML), an advanced glycation end product, is associated with proinflammation. We assessed whether serum CML and autoantibodies predicted type 1 diabetes and to what extent they were determined by genetic or environmental factors. Of 7,287 unselected schoolchildren screened, 115 were ICA(+) and were tested for baseline CML and diabetes autoantibodies and followed (for median 7 years), whereas a random selection (n = 2,102) had CML tested. CML and diabetes autoantibodies were determined in a classic twin study of twin pairs discordant for type 1 diabetes (32 monozygotic, 32 dizygotic pairs). CML was determined by enzyme-linked immunosorbent assay, autoantibodies were determined by radioimmunoprecipitation, ICA was determined by indirect immunofluorescence, and HLA class II genotyping was determined by sequence-specific oligonucleotides. CML was increased in ICA(+) and prediabetic schoolchildren and in diabetic and nondiabetic twins (all P < 0.001). Elevated levels of CML in ICA(+) children were a persistent, independent predictor of diabetes progression, in addition to autoantibodies and HLA risk. In twins model fitting, familial environment explained 75% of CML variance, and nonshared environment explained all autoantibody variance. Serum CML, a glycotoxin, emerged as an environmentally determined diabetes risk factor, in addition to autoimmunity and HLA genetic risk, and a potential therapeutic target.
Collapse
Affiliation(s)
- Huriya Beyan
- Centre for Diabetes and Metabolic Medicine, Blizard Institute, Queen Mary, University of London, London, U.K
| | - Harriette Riese
- Unit of Genetic Epidemiology & Bioinformatics, Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Interdisciplinary Center for Psychiatric Epidemiology, Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mohammed I. Hawa
- Centre for Diabetes and Metabolic Medicine, Blizard Institute, Queen Mary, University of London, London, U.K
| | - Guisi Beretta
- Centre for Diabetes and Metabolic Medicine, Blizard Institute, Queen Mary, University of London, London, U.K
| | | | - John C. Hutton
- Barbara Davis Center, University of Colorado Denver, Aurora, Colorado
| | - Huibert Burger
- Interdisciplinary Center for Psychiatric Epidemiology, Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Michael Schlosser
- Institute of Pathophysiology, Ernst Moritz Arndt University of Greifswald, Greifswald, Germany
- Division of Endocrinology and Diabetes, University Medical Center Ulm, Ulm, Germany
| | - Harold Snieder
- Unit of Genetic Epidemiology & Bioinformatics, Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bernhard O. Boehm
- Division of Endocrinology and Diabetes, University Medical Center Ulm, Ulm, Germany
| | - R. David Leslie
- Centre for Diabetes and Metabolic Medicine, Blizard Institute, Queen Mary, University of London, London, U.K
- Corresponding author: R. David Leslie,
| |
Collapse
|
50
|
Regulation of RAGE for attenuating progression of diabetic vascular complications. EXPERIMENTAL DIABETES RESEARCH 2011; 2012:894605. [PMID: 22110482 PMCID: PMC3205669 DOI: 10.1155/2012/894605] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 08/08/2011] [Accepted: 08/16/2011] [Indexed: 12/16/2022]
Abstract
Diabetic angiopathy including micro- and macroangiopathy is concerned with high rate of morbidity and mortality in patients with long-standing diabetes. Receptor for advanced glycation end products (RAGE) and its ligands have been considered as important pathogenic triggers for the progression of the vascular injuries in diabetes. The deleterious link between RAGE and diabetic angiopathy has been demonstrated in animal studies. Preventive and therapeutic strategies focusing on RAGE and its ligand axis may be of great importance in relieving diabetic vascular complications and reducing the burden of disease.
Collapse
|