51
|
Wu Y, Dong L, Song Y, Wu Y, Zhang Y, Wang S. Preventive effects of polysaccharides from Physalis alkekengi L. on dietary advanced glycation end product-induced insulin resistance in mice associated with the modulation of gut microbiota. Int J Biol Macromol 2022; 204:204-214. [PMID: 35108598 DOI: 10.1016/j.ijbiomac.2022.01.152] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/28/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022]
Abstract
Advanced glycation end products (AGEs) are commonly found in thermally processed foods, and long-term high AGE feeding has been reported to have negative effects on body health. In the current study, the effect of Physalis alkekengi L. fruit polysaccharide (PFP) on preventing dietary AGE-induced insulin resistance (IR) in mice was investigated. The results showed that PFP administration can significantly ameliorate hyperglycemia, dyslipidemia, and insulin resistance induced by dietary AGEs in mice. Compared to AGE-treated mice, the homeostasis model assessment for insulin resistance (HOMA-IR) index and insulin sensitivity (HOMA-IS) index of PFP-treated mice were improved significantly (p < 0.05). The levels of endotoxin and inflammatory cytokines in the liver decreased, while the levels of insulin receptor substrate-1 and insulin receptor substrate-2 in the liver increased (p < 0.05). The 16S rRNA analysis showed that PFP administration reversed the Bacteroidetes/Firmicutes ratio and reduced lipopolysaccharide generation and inflammation-related bacteria, including Desulfovibrio and Acetatifactor. In addition, PFP administration also increased short-chain fatty acid levels in feces compared to dietary AGE-treated mice. Spearman's correlation analysis showed that certain specific genera, including Alistipes and Caproiciproducens, are closely related to IR-related parameters. These findings suggest that PFP can prevent dietary AGE-induced IR by modulating the gut microbiota and increasing microbial metabolites.
Collapse
Affiliation(s)
- Yuekun Wu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lu Dong
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yujie Song
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yajing Wu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yan Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China.
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
52
|
Malik P, Kumar Mukherjee T. Immunological methods for the determination of AGE-RAGE axis generated glutathionylated and carbonylated proteins as oxidative stress markers. Methods 2022; 203:354-363. [DOI: 10.1016/j.ymeth.2022.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/30/2021] [Accepted: 01/26/2022] [Indexed: 11/15/2022] Open
|
53
|
A comprehensive review of advanced glycosylation end products and N- Nitrosamines in thermally processed meat products. Food Control 2022. [DOI: 10.1016/j.foodcont.2021.108449] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
54
|
Wu Q, Coumoul X, Grandjean P, Barouki R, Audouze K. Endocrine disrupting chemicals and COVID-19 relationships: A computational systems biology approach. ENVIRONMENT INTERNATIONAL 2021; 157:106232. [PMID: 33223326 PMCID: PMC7831776 DOI: 10.1016/j.envint.2020.106232] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/26/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Patients at high risk of severe forms of COVID-19 frequently suffer from chronic diseases, but other risk factors may also play a role. Environmental stressors, such as endocrine disrupting chemicals (EDCs), can contribute to certain chronic diseases and might aggravate the course of COVID-19. OBJECTIVES To explore putative links between EDCs and COVID-19 severity, an integrative systems biology approach was constructed and applied. METHODS As a first step, relevant data sets were compiled from major data sources. Biological associations of major EDCs to proteins were extracted from the CompTox database. Associations between proteins and diseases known as important COVID-19 comorbidities were obtained from the GeneCards and DisGeNET databases. Based on these data, we developed a tripartite network (EDCs-proteins-diseases) and used it to identify proteins overlapping between the EDCs and the diseases. Signaling pathways for common proteins were then investigated by over-representation analysis. RESULTS We found several statistically significant pathways that may be dysregulated by EDCs and that may also be involved in COVID-19 severity. The Th17 and the AGE/RAGE signaling pathways were particularly promising. CONCLUSIONS Pathways were identified as possible targets of EDCs and as contributors to COVID-19 severity, thereby highlighting possible links between exposure to environmental chemicals and disease development. This study also documents the application of computational systems biology methods as a relevant approach to increase the understanding of molecular mechanisms linking EDCs and human diseases, thereby contributing to toxicology prediction.
Collapse
Affiliation(s)
- Qier Wu
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Xavier Coumoul
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Philippe Grandjean
- Harvard T.H.Chan School of Public Health, Boston, MA 02115, USA; University of Southern Denmark, 5000 Odense C, Denmark
| | - Robert Barouki
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Karine Audouze
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France.
| |
Collapse
|
55
|
Diet-Derived Advanced Glycation End Products (dAGEs) Induce Proinflammatory Cytokine Expression in Cardiac and Renal Tissues of Experimental Mice: Protective Effect of Curcumin. Cardiovasc Toxicol 2021; 22:35-51. [PMID: 34655413 DOI: 10.1007/s12012-021-09697-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/17/2021] [Indexed: 10/20/2022]
Abstract
The beneficial effect of curcumin (CU) on dietary AGEs (dAGEs) involves blocking the overexpression of proinflammatory cytokine genes in the heart and kidney tissues of experimental mice. The animals were divided into six groups (n = 6/group) and were fed a heat-exposed diet (dAGEs) with or without CU for 6 months. Their blood pressure (BP) was monitored by a computerized tail-cuff BP-monitoring system. The mRNA and protein expression levels of proinflammatory genes were analyzed by RT-PCR and western blot, respectively. A marked increase in BP (108 ± 12 mmHg vs 149 ± 15 mmHg) accompanied by a marked increase in the heart and kidney weight ratio was noted in the dAGE-fed mice. Furthermore, the plasma levels of proinflammatory molecules (C5a, ICAM-1, IL-6, MCP-1, IL-1β and TNF-α) were found to be elevated (3-fold) in dAGE-fed mice. mRNA expression analysis revealed a significant increase in the expression levels of inflammatory markers (Cox-2, iNOS, and NF-κB) (3-fold) in cardiac and renal tissues of dAGE-fed mice. Moreover, increased expression of RAGE and downregulation of AGER-1 (p < 0.001) were noticed in the heart and kidney tissues of dAGE-fed mice. Interestingly, the dAGE-induced proinflammatory genes and inflammatory responses were neutralized upon cotreatment with CU. The present study demonstrates that dietary supplementation with CU has the ability to neutralize dAGE-induced adverse effects and alleviate proinflammatory gene expression in the heart and kidney tissues of experimental mice.
Collapse
|
56
|
Mahmoudinezhad M, Farhangi MA, Kahroba H, Dehghan P. Personalized diet study of dietary advanced glycation end products (AGEs) and fatty acid desaturase 2 (FADS 2) genotypes in obesity. Sci Rep 2021; 11:19725. [PMID: 34611217 PMCID: PMC8492634 DOI: 10.1038/s41598-021-99077-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023] Open
Abstract
Obesity prevalence have tripled in the past decades. It is logical to consider new approaches to halt its prevalence. In this concept, considering the effect of interaction between fatty acid desaturase 2 (FADS2) gene variants and dietary advanced glycation end products (AGEs) on obesity-related characteristics seems to be challenging. The present cross-sectional study conducted among 347 obese individuals. A validated semi-quantitative 147-item food frequency questionnaire (FFQ) was used to estimate dietary intakes and American multiethnic database was used to calculate AGEs content of food items which were not available in Iranian Food Composition Table (FCT). FADS2 gene variants were determined according to Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Analysis of covariance (ANCOVA) was used to evaluate the modifier effect of FADS2 gene-dietary AGEs on biochemical values. Based on our findings, no significant differences was reported in term of biochemical variables between AGEs tertiles. In contrast, percent of macronutrients (carbohydrate, protein and fat) of total calorie intake, amount of daily intake of fiber and meat groups showed a significant differences among AGEs tertiles. Furthermore, statistical assays clarified the modifier effects of FADS2 gene-AGEs on weight (Pinteraction = 0.04), fat mass (Pinteraction = 0.03), waist circumference (Pinteraction = 0.008) and cholesterol (Pinteraction = 0.04) level. Accordingly, higher consumption of protein or fat based foods constitute high amount of AGEs and heterozygote genotype for FADS2 tended to show lower level of AGEs content. These findings address further investigation to develop new approaches for nutritional interventions.
Collapse
Affiliation(s)
- Mahsa Mahmoudinezhad
- grid.412888.f0000 0001 2174 8913Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Attar-Neishabouri Ave, Golgasht St, 5165665931 Tabriz, Iran
| | - Mahdieh Abbasalizad Farhangi
- grid.412888.f0000 0001 2174 8913Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Houman Kahroba
- grid.412888.f0000 0001 2174 8913Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Dehghan
- grid.412888.f0000 0001 2174 8913Department of Biochemistry and Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Attar-Neishabouri Ave, Golgasht St, 5165665931 Tabriz, Iran
| |
Collapse
|
57
|
Cao C, Tang M, Zhao N, Dong S, Wu H. Effects of fish protein with glycation extent on gut microbiota and colonic barrier function in mice fed a high-fat diet. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
58
|
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: 15] [Impact Index Per Article: 5.0] [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
|
59
|
Acute Valproate Exposure Induces Mitochondrial Biogenesis and Autophagy with FOXO3a Modulation in SH-SY5Y Cells. Cells 2021; 10:cells10102522. [PMID: 34685502 PMCID: PMC8533738 DOI: 10.3390/cells10102522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/06/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Valproic acid (VPA) is an antiepileptic drug found to induce mitochondrial dysfunction and autophagy in cancer cell lines. We treated the SH-SY5Y cell line with various concentrations of VPA (1, 5, and 10 mM). The treatment decreased cell viability, ATP production, and mitochondrial membrane potential and increased reactive oxygen species production. In addition, the mitochondrial DNA copy number increased after VPA treatment in a dose-dependent manner. Western blotting showed that the levels of mitochondrial biogenesis-related proteins (PGC-1α, TFAM, and COX4) increased, though estrogen-related receptor expression decreased after VPA treatment. Further, VPA treatment increased the total and acetylated FOXO3a protein levels. Although SIRT1 expression was decreased, SIRT3 expression was increased, which regulated FOXO3 acetylation in the mitochondria. Furthermore, VPA treatment induced autophagy via increased LC3-II levels and decreased p62 expression and mTOR phosphorylation. We suggest that VPA treatment induces mitochondrial biogenesis and autophagy via changes in FOXO3a expression and posttranslational modification in the SH-SY5Y cell line.
Collapse
|
60
|
Cordova R, Kliemann N, Huybrechts I, Rauber F, Vamos EP, Levy RB, Wagner KH, Viallon V, Casagrande C, Nicolas G, Dahm CC, Zhang J, Halkjær J, Tjønneland A, Boutron-Ruault MC, Mancini FR, Laouali N, Katzke V, Srour B, Jannasch F, Schulze MB, Masala G, Grioni S, Panico S, van der Schouw YT, Derksen JWG, Rylander C, Skeie G, Jakszyn P, Rodriguez-Barranco M, Huerta JM, Barricarte A, Brunkwall L, Ramne S, Bodén S, Perez-Cornago A, Heath AK, Vineis P, Weiderpass E, Monteiro CA, Gunter MJ, Millett C, Freisling H. Consumption of ultra-processed foods associated with weight gain and obesity in adults: A multi-national cohort study. Clin Nutr 2021; 40:5079-5088. [PMID: 34455267 DOI: 10.1016/j.clnu.2021.08.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/02/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND There is a worldwide shift towards increased consumption of ultra-processed foods (UPF) with concurrent rising prevalence of obesity. We examined the relationship between the consumption of UPF and weight gain and risk of obesity. METHODS This prospective cohort included 348 748 men and women aged 25-70 years. Participants were recruited between 1992 and 2000 from 9 European countries in the European Prospective Investigation into Cancer and Nutrition (EPIC) study. Two body weight measures were available, at baseline and after a median follow-up time of 5 years. Foods and drinks were assessed at baseline by dietary questionnaires and classified according to their degree of processing using NOVA classification. Multilevel mixed linear regression was used to estimate the association between UPF consumption and body weight change (kg/5 years). To estimate the relative risk of becoming overweight or obese after 5 years we used Poisson regression stratified according to baseline body mass index (BMI). RESULTS After multivariable adjustment, higher UPF consumption (per 1 SD increment) was positively associated with weight gain (0·12 kg/5 years, 95% CI 0·09 to 0·15). Comparing highest vs. lowest quintile of UPF consumption was associated with a 15% greater risk (95% CI 1·11, 1·19) of becoming overweight or obese in normal weight participants, and with a 16% greater risk (95% CI 1·09, 1·23) of becoming obese in participants who were overweight at baseline. CONCLUSIONS These results are supportive of public health campaigns to substitute UPF for less processed alternatives for obesity prevention and weight management.
Collapse
Affiliation(s)
- Reynalda Cordova
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France; Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Nathalie Kliemann
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Inge Huybrechts
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Fernanda Rauber
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, Brazil; Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Eszter P Vamos
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, UK
| | - Renata Bertazzi Levy
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, Brazil; Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Vivian Viallon
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Corinne Casagrande
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Geneviève Nicolas
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | | | - Jie Zhang
- Department of Public Health, Aarhus University, Denmark
| | - Jytte Halkjær
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Christine Boutron-Ruault
- French National Institute of Health and Medical Research (INSERM), Centre for Research in Epidemiology and Population Health (CESP), UVSQ, Université Paris-Saclay, Université Paris-Sud, France; Institut Gustave Roussy, F-94805, Villejuif, France
| | - Francesca Romana Mancini
- French National Institute of Health and Medical Research (INSERM), Centre for Research in Epidemiology and Population Health (CESP), UVSQ, Université Paris-Saclay, Université Paris-Sud, France; Institut Gustave Roussy, F-94805, Villejuif, France
| | - Nasser Laouali
- French National Institute of Health and Medical Research (INSERM), Centre for Research in Epidemiology and Population Health (CESP), UVSQ, Université Paris-Saclay, Université Paris-Sud, France; Institut Gustave Roussy, F-94805, Villejuif, France
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernard Srour
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Franziska Jannasch
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; NutriAct - Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Giovanna Masala
- Molecular and Lifestyle Epidemiology Branch, Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network ISPRO, Florence, Italy
| | - Sara Grioni
- Epidemiology and Prevention Unit Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian, 120133, Milano, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica E Chirurgia Federico II University, Naples, Italy
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jeroen W G Derksen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Charlotta Rylander
- Department of Community Medicine, UiT the Arctic University of Norway, Tromsø, Norway
| | - Guri Skeie
- Department of Community Medicine, UiT the Arctic University of Norway, Tromsø, Norway
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Barcelona, Spain; Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Miguel Rodriguez-Barranco
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - José María Huerta
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Murcia, Spain
| | - Aurelio Barricarte
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Navarra Public Health Institute, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | | | - Stina Ramne
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Stina Bodén
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, UK
| | - Alicia K Heath
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, UK
| | - Paolo Vineis
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, UK
| | - Elisabete Weiderpass
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Carlos Augusto Monteiro
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, Brazil; Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Christopher Millett
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, UK
| | - Heinz Freisling
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France.
| |
Collapse
|
61
|
Wouters K, Cento AS, Gaens KH, Teunissen M, Scheijen JLJM, Barutta F, Chiazza F, Collotta D, Aragno M, Gruden G, Collino M, Schalkwijk CG, Mastrocola R. Deletion of RAGE fails to prevent hepatosteatosis in obese mice due to impairment of other AGEs receptors and detoxifying systems. Sci Rep 2021; 11:17373. [PMID: 34462492 PMCID: PMC8405685 DOI: 10.1038/s41598-021-96859-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Advanced glycation endproducts (AGEs) are involved in several diseases, including NAFLD and NASH. RAGE is the main receptor mediating the pro-inflammatory signalling induced by AGEs. Therefore, targeting of RAGE has been proposed for prevention of chronic inflammatory diseases. However, the role of RAGE in the development of NAFLD and NASH remains poorly understood. We thus aimed to analyse the effect of obesity on AGEs accumulation, AGE-receptors and AGE-detoxification, and whether the absence of RAGE might improve hepatosteatosis and inflammation, by comparing the liver of lean control, obese (LeptrDb-/-) and obese RAGE-deficient (RAGE-/- LeptrDb-/-) mice. Obesity induced AGEs accumulation and RAGE expression with hepatosteatosis and inflammation in LeptrDb-/-, compared to lean controls. Despite the genetic deletion of RAGE in the LeptrDb-/- mice, high levels of intrahepatic AGEs were maintained accompanied by decreased expression of the protective AGE-receptor-1, impaired AGE-detoxifying system glyoxalase-1, and increased expression of the alternative AGE-receptor galectin-3. We also found sustained hepatosteatosis and inflammation as determined by persistent activation of the lipogenic SREBP1c and proinflammatory NLRP3 signalling pathways. Thus, RAGE targeting is not effective in the prevention of NAFLD in conditions of obesity, likely due to the direct liver specific crosstalk of RAGE with other AGE-receptors and AGE-detoxifying systems.
Collapse
Affiliation(s)
- Kristiaan Wouters
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Alessia S. Cento
- grid.7605.40000 0001 2336 6580Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Katrien H. Gaens
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Margee Teunissen
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands
| | - Jean L. J. M. Scheijen
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Federica Barutta
- grid.7605.40000 0001 2336 6580Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fausto Chiazza
- grid.16563.370000000121663741Department of Drug Sciences, University of Eastern Piedmont, Novara, Italy
| | - Debora Collotta
- grid.7605.40000 0001 2336 6580Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Manuela Aragno
- grid.7605.40000 0001 2336 6580Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Gabriella Gruden
- grid.7605.40000 0001 2336 6580Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Collino
- grid.7605.40000 0001 2336 6580Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Casper G. Schalkwijk
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.5012.60000 0001 0481 6099Cardiovascular Research Institute Maastricht, Maastricht, Limburg The Netherlands
| | - Raffaella Mastrocola
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, MUMC, Maastricht, Limburg The Netherlands ,grid.7605.40000 0001 2336 6580Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| |
Collapse
|
62
|
Garay-Sevilla ME, Rojas A, Portero-Otin M, Uribarri J. Dietary AGEs as Exogenous Boosters of Inflammation. Nutrients 2021; 13:nu13082802. [PMID: 34444961 PMCID: PMC8401706 DOI: 10.3390/nu13082802] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Most chronic modern non-transmissible diseases seem to begin as the result of low-grade inflammation extending over prolonged periods of time. The importance of diet as a source of many pro-inflammatory compounds that could create and sustain such a low-grade inflammatory state cannot be ignored, particularly since we are constantly exposed to them during the day. The focus of this review is on specific components of the diet associated with inflammation, specifically advanced glycation end products (AGEs) that form during thermal processing of food. AGEs are also generated in the body in normal physiology and are widely recognized as increased in diabetes, but many people are unaware of the potential importance of exogenous AGEs ingested in food. We review experimental models, epidemiologic data, and small clinical trials that suggest an important association between dietary intake of these compounds and development of an inflammatory and pro-oxidative state that is conducive to chronic diseases. We compare dietary intake of AGEs with other widely known dietary patterns, such as the Mediterranean and the Dietary Approaches to Stop Hypertension (DASH) diets, as well as the Dietary Inflammation Index (DII). Finally, we delineate in detail the pathophysiological mechanisms induced by dietary AGEs, both direct (i.e., non-receptor-mediated) and indirect (receptor-mediated).
Collapse
Affiliation(s)
| | - Armando Rojas
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad Catolica del Maule, Talca 3480005, Chile;
| | - Manuel Portero-Otin
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad de Lleida, 25196 Lleida, Spain;
| | - Jaime Uribarri
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence: ; Tel.: +1-212-241-1887
| |
Collapse
|
63
|
Yu W, Fan L, Wang M, Cao B, Hu X. Pterostilbene Improves Insulin Resistance Caused by Advanced Glycation End Products (AGEs) in Hepatocytes and Mice. Mol Nutr Food Res 2021; 65:e2100321. [PMID: 34085383 DOI: 10.1002/mnfr.202100321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/15/2021] [Indexed: 11/07/2022]
Abstract
SCOPE Increased consumption of modern processed foods rich in AGEs is drawing worldwide concerns because they are related with rising diabetes prevalence. This study aimed to investigate if pterostilbene (PTE) regulates glucose metabolism and insulin signaling, as well as its potential mechanism in the context of AGEs exposure. METHODS AND RESULTS In vitro, Lo2 and HepG2 cells are treated with vehicle, AGEs with or without PTE. AGEs exposure directly impair insulin action as evidenced by assays of insulin-stimulated glucose uptake, consumption, and output. However, PTE efficiently rescue the AGE-induced phenotypes in both cell lines, and enhance IRS-1/PI3K/AKT insulin signaling in a dose-dependent manner. In vivo, C57BL6 mice are fed with regular, high AGEs diet and high AGEs plus PTE. PTE administration effectively improves hyperglycemia, glucose tolerance, and impaired hepatic insulin signaling induced by AGEs, consistent with the in vitro experiments. Moreover, PTE reduce AGEs accumulation in liver and serum. RNA-seq data indicate that PTE counteracts several AGEs-induced dysfunctions including diabetes related process, glucose metabolic process, immune response, and so on. CONCLUSION PTE treatment prominently reduced AGEs accumulation and alleviated AGEs-associated diabetes symptoms. PTE could be used as a promising glucose-sensitizing agent for nutritional intervention.
Collapse
Affiliation(s)
- Wenzhe Yu
- School of Medicine, Xiamen University, Xiamen, 361102, P. R. China
| | - Lida Fan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Heath, Xiamen University, Xiamen, 361102, P. R. China
| | - Mingfu Wang
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, P. R. China
| | - Bin Cao
- School of Medicine, Xiamen University, Xiamen, 361102, P. R. China
| | - Xiaoqian Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Heath, Xiamen University, Xiamen, 361102, P. R. China
| |
Collapse
|
64
|
Chen Y, Guo TL. Dietary advanced glycation end-products elicit toxicological effects by disrupting gut microbiome and immune homeostasis. J Immunotoxicol 2021; 18:93-104. [PMID: 34436982 PMCID: PMC9885815 DOI: 10.1080/1547691x.2021.1959677] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The aging immune system is characterized by a low-grade chronic systemic inflammatory state ("inflammaging") marked by elevated serum levels of inflammatory molecules such as interleukin (IL)-6 and C-reactive protein (CRP). These inflammatory markers were also reported to be strong predictors for the development/severity of Type 2 diabetes, obesity, and COVID-19. The levels of these markers have been positively associated with those of advanced glycation end-products (AGEs) generated via non-enzymatic glycation and oxidation of proteins and lipids during normal aging and metabolism. Based on the above observations, it is clinically important to elucidate how dietary AGEs modulate inflammation and might thus increase the risk for aging-exacerbated diseases. The present narrative review discusses the potential pro-inflammatory properties of dietary AGEs with a focus on the inflammatory mediators CRP, IL-6 and ferritin, and their relations to aging in general and Type 2 diabetes in particular. In addition, underlying mechanisms - including those related to gut microbiota and the receptors for AGEs, and the roles AGEs might play in affecting physiologies of the healthy elderly, obese individuals, and diabetics are discussed in regard to any greater susceptibility to COVID-19.
Collapse
Affiliation(s)
- Yingjia Chen
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Tai L. Guo
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
65
|
Hydroxytyrosol Selectively Affects Non-Enzymatic Glycation in Human Insulin and Protects by AGEs Cytotoxicity. Antioxidants (Basel) 2021; 10:antiox10071127. [PMID: 34356360 PMCID: PMC8301023 DOI: 10.3390/antiox10071127] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Hydroxytyrosol (HT), the major phenolic compound in olive oil, is attracting increasing interest for its beneficial properties including a notable antioxidant and anti-inflammatory power. In this study, using a combination of biophysical and cell biology techniques, we have tested the role of HT in the formation of advanced glycation end-products (AGEs). AGEs have a key role in clinical sciences as they have been associated to diabetes, neurodegenerative and cardiovascular diseases. In addition, as the incidence of Alzheimer’s disease (AD) is strongly increased in diabetic patients, AGE formation is supposed to be involved in the development of the pathological hallmarks of AD. Our data show that HT selectively inhibits protein glycation reaction in human insulin, and it is able to counteract the AGE-induced cytotoxicity in human neurotypical cells by acting on SIRT1 level and oxidative stress, as well as on inflammatory response. This study identifies new beneficial properties for HT and suggests it might be a promising molecule in protecting against the AGE-induced toxicity, a key mechanism underlying the development and progression of neurodegenerative disorders.
Collapse
|
66
|
Dietary advanced glycation endproducts (AGEs) increase their concentration in plasma and tissues, result in inflammation and modulate gut microbial composition in mice; evidence for reversibility. Food Res Int 2021; 147:110547. [PMID: 34399524 DOI: 10.1016/j.foodres.2021.110547] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/04/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022]
Abstract
SCOPE Dietary advanced glycation endproducts (AGEs) are associated with negative biological effects, possibly due to accumulation in plasma and tissues and through modulation of inflammation and gut microbiota. Whether these biological consequences are reversible by limiting dietary AGE intake is unknown. METHODS AND RESULTS Young healthy C57BL/6 mice were fed a standard chow (n = 10) or a baked chow high AGE-diet (n = 10) (~1.8-6.9 fold increased protein-bound Nε-(carboxymethyl)lysine (CML), Nε-(1-carboxyethyl)lysine (CEL), and Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1)) for 10 weeks or a switch diet with baked chow for 5 weeks followed by 5 weeks of standard chow (n = 10). We assessed accumulation of AGEs in plasma, kidney, and liver and measured inflammatory markers and gut microbial composition. After 10 weeks of baked chow, a substantial panel of AGEs were increased in plasma, liver, and kidney. These increases were normalized after the switch diet. The inflammatory z-score increased after the baked chow diet. Gut microbial composition differed significantly between groups, with enriched Dubosiella spp. dominating these alterations. CONCLUSION A high AGE-diet led to an increase of AGEs in plasma, kidney, and liver and to more inflammation and modification of the gut microbiota. These effects were reversed or discontinued by a diet lower in AGEs.
Collapse
|
67
|
Byrne NJ, Rajasekaran NS, Abel ED, Bugger H. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radic Biol Med 2021; 169:317-342. [PMID: 33910093 PMCID: PMC8285002 DOI: 10.1016/j.freeradbiomed.2021.03.046] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Even in the absence of coronary artery disease and hypertension, diabetes mellitus (DM) may increase the risk for heart failure development. This risk evolves from functional and structural alterations induced by diabetes in the heart, a cardiac entity termed diabetic cardiomyopathy (DbCM). Oxidative stress, defined as the imbalance of reactive oxygen species (ROS) has been increasingly proposed to contribute to the development of DbCM. There are several sources of ROS production including the mitochondria, NAD(P)H oxidase, xanthine oxidase, and uncoupled nitric oxide synthase. Overproduction of ROS in DbCM is thought to be counterbalanced by elevated antioxidant defense enzymes such as catalase and superoxide dismutase. Excess ROS in the cardiomyocyte results in further ROS production, mitochondrial DNA damage, lipid peroxidation, post-translational modifications of proteins and ultimately cell death and cardiac dysfunction. Furthermore, ROS modulates transcription factors responsible for expression of antioxidant enzymes. Lastly, evidence exists that several pharmacological agents may convey cardiovascular benefit by antioxidant mechanisms. As such, increasing our understanding of the pathways that lead to increased ROS production and impaired antioxidant defense may enable the development of therapeutic strategies against the progression of DbCM. Herein, we review the current knowledge about causes and consequences of ROS in DbCM, as well as the therapeutic potential and strategies of targeting oxidative stress in the diabetic heart.
Collapse
Affiliation(s)
- Nikole J Byrne
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
68
|
Giandalia A, Giuffrida AE, Gembillo G, Cucinotta D, Squadrito G, Santoro D, Russo GT. Gender Differences in Diabetic Kidney Disease: Focus on Hormonal, Genetic and Clinical Factors. Int J Mol Sci 2021; 22:5808. [PMID: 34071671 PMCID: PMC8198374 DOI: 10.3390/ijms22115808] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the most serious complications of both type 1 (T1DM) and type 2 diabetes mellitus (T2DM). Current guidelines recommend a personalized approach in order to reduce the burden of DM and its complications. Recognizing sex and gender- differences in medicine is considered one of the first steps toward personalized medicine, but the gender issue in DM has been scarcely explored so far. Gender differences have been reported in the incidence and the prevalence of DKD, in its phenotypes and clinical manifestations, as well as in several risk factors, with a different impact in the two genders. Hormonal factors, especially estrogen loss, play a significant role in explaining these differences. Additionally, the impact of sex chromosomes as well as the influence of gene-sex interactions with several susceptibility genes for DKD have been investigated. In spite of the increasing evidence that sex and gender should be included in the evaluation of DKD, several open issues remain uncovered, including the potentially different effects of newly recommended drugs, such as SGLT2i and GLP1Ras. This narrative review explored current evidence on sex/gender differences in DKD, taking into account hormonal, genetic and clinical factors.
Collapse
Affiliation(s)
- Annalisa Giandalia
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.G.); (D.C.); (G.S.)
| | - Alfio Edoardo Giuffrida
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (G.G.); (D.S.)
| | - Guido Gembillo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (G.G.); (D.S.)
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy
| | - Domenico Cucinotta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.G.); (D.C.); (G.S.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.G.); (D.C.); (G.S.)
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (G.G.); (D.S.)
| | - Giuseppina T. Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.G.); (D.C.); (G.S.)
| |
Collapse
|
69
|
Yue Y, Wang J, Shen P, Kim KH, Park Y. Methylglyoxal influences development of Caenorhabditis elegans via lin-41-dependent pathway. Food Chem Toxicol 2021; 152:112238. [PMID: 33901606 DOI: 10.1016/j.fct.2021.112238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 01/02/2023]
Abstract
Methylglyoxal is a highly reactive dicarbonyl compound. It can be obtained either endogenously through biological enzymatic/non-enzymatic pathways or exogenously via the uptake of certain foods and beverages, such as Manuka honey. Studies about its biological properties are quite controversial, though the majority reported a positive association between methylglyoxal and certain pathologies. In this report, we tested if methylglyoxal can alter the development of animals using Caenorhabditis elegans as the in vivo model. Treatment of methylglyoxal at 0.1 and 1 mmol/L for 2 days significantly inhibited the development of Caenorhabditis elegans, particularly targeting the transition from L3 stage. Pharyngeal pumping rate, the food intake marker was also significantly reduced by methylglyoxal at both 0.1 and 1 mmol/L. Additionally, treatment of 0.1 mmol/L methylglyoxal increased, while 1 mmol/L methylglyoxal decreased the nematodes' average moving speed. The effect of methylglyoxal on development was in part due to the modulation of lin-41, which encodes a homolog of human TRIM71. The mutation of lin-41 could alleviate or abolish the effects of methylglyoxal on growth rate, body size, pumping rate and locomotive activity. In summary, these results suggested that methylglyoxal influenced the development of Caenorhabditis elegans, which is in part via the lin-41-dependent pathway.
Collapse
Affiliation(s)
- Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| | - Jiaying Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| | - Peiyi Shen
- Department of Agriculture, Culinology® and Hospitality Management, Southwest Minnesota State University, Marshall, MN, 56258, USA
| | - Kee-Hong Kim
- Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
70
|
Zhang Y, Dong L, Zhang J, Shi J, Wang Y, Wang S. Adverse Effects of Thermal Food Processing on the Structural, Nutritional, and Biological Properties of Proteins. Annu Rev Food Sci Technol 2021; 12:259-286. [PMID: 33770470 DOI: 10.1146/annurev-food-062320-012215] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Thermal processing is one of the most important processing methods in the food industry. However, many studies have revealed that thermal processing can have detrimental effects on the nutritional and functional properties of foods because of the complex interactions among food components. Proteins are essential nutrients for humans, and changes in the structure and nutritional properties of proteins can substantially impact the biological effects of foods. This review focuses on the interactions among proteins, sugars, and lipids during thermal food processing and the effects of these interactions on the structure, nutritional value, and biological effects of proteins. In particular, the negative effects of modified proteins on human health and strategies for mitigating these detrimental effects from two perspectives, namely, reducing the formation of modified proteins during thermal processing and dietary intervention in vivo, are discussed.
Collapse
Affiliation(s)
- Yan Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| | - Lu Dong
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| | - Jinhui Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| | - Jiaqi Shi
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| | - Yaya Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| |
Collapse
|
71
|
Garay-Sevilla ME, Gomez-Ojeda A, González I, Luévano-Contreras C, Rojas A. Contribution of RAGE axis activation to the association between metabolic syndrome and cancer. Mol Cell Biochem 2021; 476:1555-1573. [PMID: 33398664 DOI: 10.1007/s11010-020-04022-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
Far beyond the compelling proofs supporting that the metabolic syndrome represents a risk factor for diabetes and cardiovascular diseases, a growing body of evidence suggests that it is also a risk factor for different types of cancer. However, the involved molecular mechanisms underlying this association are not fully understood, and they have been mainly focused on the individual contributions of each component of the metabolic syndrome such as obesity, hyperglycemia, and high blood pressure to the development of cancer. The Receptor for Advanced Glycation End-products (RAGE) axis activation has emerged as an important contributor to the pathophysiology of many clinical entities, by fueling a chronic inflammatory milieu, and thus supporting an optimal microenvironment to promote tumor growth and progression. In the present review, we intend to highlight that RAGE axis activation is a crosswise element on the potential mechanistic contributions of some relevant components of metabolic syndrome into the association with cancer.
Collapse
Affiliation(s)
- Ma Eugenia Garay-Sevilla
- Department of Medical Science, Division of Health Science, University of Guanajuato, Campus León, Guanajuato, Mexico
| | - Armando Gomez-Ojeda
- Department of Medical Science, Division of Health Science, University of Guanajuato, Campus León, Guanajuato, Mexico
| | - Ileana González
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Claudia Luévano-Contreras
- Department of Medical Science, Division of Health Science, University of Guanajuato, Campus León, Guanajuato, Mexico
| | - Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| |
Collapse
|
72
|
Quan W, Zeng M, Jiao Y, Li Y, Xue C, Liu G, Wang Z, Qin F, He Z, Chen J. Western Dietary Patterns, Foods, and Risk of Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Adv Nutr 2021; 12:1353-1364. [PMID: 33578428 PMCID: PMC8321835 DOI: 10.1093/advances/nmaa184] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/16/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
An increasing number of epidemiological studies suggest that adherence to Western dietary patterns (WDPs) is associated with risk of gestational diabetes mellitus (GDM), but results remain inconsistent. Therefore, we conducted a systematic review and meta-analysis of the effect of WDPs and typical Western dietary foods on GDM. A literature search was performed in PubMed, Embase, Web of Knowledge, and the Cochrane Library up to December 2019. Cohort studies investigating the combined associations of WDPs with incidence of GDM were included. Reviewers were paired, and they independently reviewed and assessed studies, extracted data, and evaluated study quality. Pooled HRs were calculated using random-effects models. Heterogeneity and publication bias tests were also conducted. Twenty-one prospective cohort studies with 191,589 participants, including 12,331 women with GDM, were included in our analysis. The pooled risk ratio (RR) of WDPs was 1.52 (95% CI: 1.21, 1.91), indicating a significant association with GDM risk in Western countries. Potatoes (pooled RR: 1.12; 95% CI: 0.93, 1.35) showed a nonsignificant (P > 0.05) relation to GDM risk. However, consumption of animal meat (pooled RR: 1.35; 95% CI: 1.16, 1.57) and fast food (pooled RR: 1.75; 95% CI: 1.41, 2.19) showed a positive association with the risk of developing GDM. Subgroup analysis demonstrated that the consumption of red meat and processed red meat increased the risk of GDM more than either poultry or fish intake. Our study provides further evidence for understanding the relation between dietary factors and increased GDM risk and contributes to reducing the incidence of GDM through healthy diets.
Collapse
Affiliation(s)
- Wei Quan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ye Jiao
- School of Chemistry and Food Engineering, Changsha University of Science & Technology, Changsha, China
| | - Yong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chaoyi Xue
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Guoping Liu
- Wuxi People's Hospital, Nanjing Medical University, Wuxi, China
| | - Zhaojun Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Fang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | | | - Jie Chen
- Address correspondence to JC (e-mail: )
| |
Collapse
|
73
|
From mitochondria to sarcopenia: Role of inflammaging and RAGE-ligand axis implication. Exp Gerontol 2021; 146:111247. [PMID: 33484891 DOI: 10.1016/j.exger.2021.111247] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Sarcopenia is characterized by a loss of muscle mass and function that reduces mobility, diminishes quality of life, and can lead to fall-related injuries. At the intracellular level, mitochondrial population alterations are considered as key contributors to the complex etiology of sarcopenia. Mitochondrial dysfunctions lead to reactive oxygen species production, altered cellular proteostasis, and promotes inflammation. Interestingly, the receptor for advanced glycation end-products (RAGE) is a pro-inflammatory receptor involved in inflammaging. In this review, after a brief description of sarcopenia, we will describe how mitochondria and the pathways controlling mitochondrial population quality could participate to age-induced muscle mass and force loss. Finally, we will discuss the RAGE-ligand axis during aging and its possible connection with mitochondria to control inflammaging and sarcopenia.
Collapse
|
74
|
García-Gómez E, Bobadilla-Bravo M, Díaz-Díaz E, Vázquez-Martínez ER, Nava-Salazar S, Torres-Ramos Y, García-Romero CS, Camacho-Arroyo I, Cerbón M. High Plasmatic Levels of Advanced Glycation End Products are Associated with Metabolic Alterations and Insulin Resistance in Preeclamptic Women. Curr Mol Med 2021; 20:751-759. [DOI: 10.2174/1566524020666200220141414] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 11/22/2022]
Abstract
Aims:
The purpose of this study was to investigate the association between
plasmatic levels of advanced end glycation products (AGEs) and the metabolic profile in
subjects diagnosed with preeclampsia, due to the known relation of these molecules with
oxidative stress and inflammation, which in turn are related with PE pathogenesis.
Background:
It has been reported that increased levels of AGEs are observed in patients
with preeclampsia as compared with healthy pregnant subjects, which was mainly
associated with oxidative stress and inflammation. Besides, in women with preeclampsia,
there are metabolic changes such as hyperinsulinemia, glucose intolerance, dyslipidemia,
among others, that are associated with an exacerbated insulin resistance. Additionally,
some parameters indicate the alteration of hepatic function, such as increased levels of liver
enzymes. However, the relationship of levels of AGEs with altered lipidic, hepatic, and
glucose metabolism parameters in preeclampsia has not been evaluated.
Objective:
To investigate the association between plasmatic levels of AGEs and hepatic,
lipid, and metabolic profiles in women diagnosed with preeclampsia.
Methods:
Plasma levels of AGEs were determined by a competitive enzyme-linked
immunosorbent assay (ELISA) in 15 patients diagnosed with preeclampsia and 28
normoevolutive pregnant subjects (control group). Hepatic (serum creatinine, gammaglutamyl
transpeptidase, aspartate transaminase, alanine transaminase, uric acid, and
lactate dehydrogenase), lipid (apolipoprotein A, apolipoprotein B, total cholesterol,
triglycerides, low-density lipoproteins, and high-density lipoproteins), and metabolic
variables (glucose, insulin, and insulin resistance) were assessed.
Results:
Plasmatic levels of AGEs were significantly higher in patients with preeclampsia
as compared with the control. A positive correlation between circulating levels of AGEs and
gamma-glutamyl transpeptidase, uric acid, glucose, insulin, and HOMA-IR levels was found
in patients with preeclampsia. In conclusion, circulating levels of AGEs were higher in
patients with preeclampsia than those observed in healthy pregnant subjects. Besides,
variables of hepatic and metabolic profile, particularly those related to insulin resistance,
were higher in preeclampsia as compared with healthy pregnant subjects. Interestingly,
there is a positive correlation between AGEs levels and insulin resistance.
Conclusions:
Circulating levels of AGEs were higher in patients with preeclampsia than
those observed in healthy pregnant subjects. Besides, hepatic and metabolic profiles,
particularly those related to insulin resistance, were higher in preeclampsia as compared
with healthy pregnant subjects. Interestingly, there is a positive correlation between AGEs
levels and insulin resistance, suggesting that excessive glycation and an impaired
metabolic profile contribute to the physiopathology of preeclampsia.
Collapse
Affiliation(s)
- Elizabeth García-Gómez
- Unidad de Investigacion en Reproduccion Humana, Consejo Nacional de Ciencia y Tecnologia (CONACyT)- Instituto Nacional de Perinatologia, Mexico
| | - Mariana Bobadilla-Bravo
- Unidad de Investigacion en Reproduccion Humana, Instituto Nacional de Perinatologia-Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Mexico
| | - Eulises Díaz-Díaz
- Departamento de Biologia de la Reproduccion, Instituto Nacional de Ciencias Medicas y Nutricion “Salvador Zubiran”, Mexico
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigacion en Reproduccion Humana, Instituto Nacional de Perinatologia-Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Mexico
| | - Sonia Nava-Salazar
- Departamento de Inmunobioquimica, Instituto Nacional de Perinatologia "Isidro Espinosa de los Reyes, Mexico
| | - Yessica Torres-Ramos
- Departamento de Inmunobioquimica, Instituto Nacional de Perinatologia "Isidro Espinosa de los Reyes, Mexico
| | - Carmen Selene García-Romero
- Departamento de Infectologia e Inmunologia, Instituto Nacional de Perinatologia "Isidro Espinosa de los Reyes", Ciudad de Mexico, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigacion en Reproduccion Humana, Instituto Nacional de Perinatologia-Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Mexico
| | - Marco Cerbón
- Unidad de Investigacion en Reproduccion Humana, Instituto Nacional de Perinatologia-Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Mexico
| |
Collapse
|
75
|
Kshirsagar V, Thingore C, Juvekar A. Insulin resistance: a connecting link between Alzheimer's disease and metabolic disorder. Metab Brain Dis 2021; 36:67-83. [PMID: 32986168 DOI: 10.1007/s11011-020-00622-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Recent evidence suggests that Alzheimer's disease (AD) is closely linked with insulin resistance, as seen in type 2 diabetes mellitus (T2DM). Insulin signaling is impaired in AD brains due to insulin resistance, ultimately resulting in the formation of neurofibrillary tangles (NFTs). AD and T2DM are connected at molecular, clinical, and epidemiological levels making it imperative to understand the contribution of T2DM, and other metabolic disorders, to AD pathogenesis. In this review, we have discussed various modalities involved in the pathogenesis of these two diseases and explained the contributing parameters. Insulin is vital for maintaining glucose homeostasis and it plays an important role in regulating inflammation. Here, we have discussed the roles of various contributing factors like miRNA, leptin hormone, neuroinflammation, metabolic dysfunction, and gangliosides in insulin impairment both in AD and T2DM. Understanding these mechanisms will be a big step forward for making molecular therapies that may help maintain or prevent both AD and T2DM, thus reducing the burden of both these diseases.
Collapse
Affiliation(s)
- Viplav Kshirsagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India
| | - Chetan Thingore
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India
| | - Archana Juvekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India.
| |
Collapse
|
76
|
Garay-Sevilla ME, Beeri MS, de la Maza MP, Rojas A, Salazar-Villanea S, Uribarri J. The potential role of dietary advanced glycation endproducts in the development of chronic non-infectious diseases: a narrative review. Nutr Res Rev 2020; 33:298-311. [PMID: 32238213 DOI: 10.1017/s0954422420000104] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing clinical and experimental evidence accumulated during the past few decades supports an important role for dietary advanced glycation endproducts (AGE) in the pathogenesis of many chronic non-infectious diseases, such as type 2 diabetes, CVD and others, that are reaching epidemic proportions in the Western world. Although AGE are compounds widely recognised as generated in excess in the body in diabetic patients, the potential importance of exogenous AGE, mostly of dietary origin, has been largely ignored in the general nutrition audience. In the present review we aim to describe dietary AGE, their mechanisms of formation and absorption into the body as well as their main mechanisms of action. We will present in detail current evidence of their potential role in the development of several chronic non-infectious clinical conditions, some general suggestions on how to restrict them in the diet and evidence regarding the potential benefits of lowering their consumption.
Collapse
Affiliation(s)
- M E Garay-Sevilla
- Medical Science Department, University of Guanajuato, Guanajuato, Mexico
| | - M S Beeri
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
| | - M P de la Maza
- Institute of Nutrition and Food Technology Dr. Fernando Monckeberg Barros, University of Chile, Santiago, Chile
| | - A Rojas
- Biomedical Research Laboratories, Faculty of Medicine, Catholic University of Maule, Talca, Chile
| | - S Salazar-Villanea
- Department of Animal Science, Universidad de Costa Rica, San Pedro Montes de Oca, San José, Costa Rica
| | - J Uribarri
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
77
|
Quan W, Jiao Y, Xue C, Li Y, Wang Z, Zeng M, Qin F, He Z, Chen J. Processed potatoes intake and risk of type 2 diabetes: a systematic review and meta-analysis of nine prospective cohort studies. Crit Rev Food Sci Nutr 2020; 62:1417-1425. [PMID: 33153277 DOI: 10.1080/10408398.2020.1843395] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The current cohort study shows the inconsistent association between potato consumption and the risk of type 2 diabetes mellitus (T2DM). Therefore, we conducted a systematic review and dose-response meta-analysis of published prospective cohort studies to quantitatively estimate this association. We searched PubMed, Embase, MEDLINE, Web of Knowledge, and the Cochrane Library up to September 2019 for all published articles. Seven of the articles reported nine cohort studies with 383,211 participants, with 23,189 T2DM cases that met the inclusion criteria and were included for our analysis. The results of random effects model pooled relative risk (RR) showed an association between potato intake and the risk of T2DM (pooled RR = 1.13, 95% CI: 1.02-1.26, p > 0.01). In the subgroup analysis, French fries, long-term follow-up, large sample size, and high-quality studies were associated with an increased T2DM risk. Further, a linear dose-response analysis indicated that 100 g/day increment of total potato (RR = 1.05, 95% CI: 1.02-1.08) and French fries (RR = 1.10, 95% CI: 1.07-1.14) consumption may increase the risk of T2DM by 5% and 10%, respectively. Our meta-analysis showed that potato consumption, especially French fries consumption, was associated with increased T2DM risk.
Collapse
Affiliation(s)
- Wei Quan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Ye Jiao
- School of Chemistry and Food Engineering, Changsha University of Science & Technology, Changsha, China
| | - Chaoyi Xue
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhaojun Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Fang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhiyong He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jie Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
78
|
Mi J, Song J, Zhao Y, Wu X. Association of hemoglobin glycation index and its interaction with obesity/family history of hypertension on hypertension risk: a community-based cross-sectional survey. BMC Cardiovasc Disord 2020; 20:477. [PMID: 33148181 PMCID: PMC7640660 DOI: 10.1186/s12872-020-01762-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/29/2020] [Indexed: 01/12/2023] Open
Abstract
Background Hemoglobin glycation index (HGI) is considered to be a convenient measurable indicator to assess the inter-individual variation of HbA1c. In the present study, we tested the relationship between HGI and risk of hypertension, and further explored the possible interacting influences of HGI with other such factors on hypertension risk among Chinese individuals. Methods The eligible subjects were chosen from a community-based cross-sectional survey in China. We collected relevant data and clinical indicators for each participant. HGI was calculated as “measured HbA1c-predicted HbA1c” and divided into four categories according to quartile. The following indicators were used to assess interactive effects: (1) relative excess risk due to interaction (RERI); (2) attributable proportion due to interaction (AP); and (3) synergy index (SI). Statistical analysis was performed using R software. Results Specifically, 1777 eligible participants were selected in this cross-sectional survey. There were 433 subjects who were identified to have hypertension (24.4%). A significant increase in the prevalence of hypertension from Q1 to Q4 of HGI was observed (p < 0.001). Multivariable logistic model demonstrated that subjects at the highest HGI group had a substantially increased risk of being hypertensive than subjects in the first quartile of HGI, as indicated by the OR value of 1.87 (95% CI 1.26–2.78). Moreover, a significant interaction between family history of hypertension and HGI on hypertension risk was detected (RERI: 1.36, 95% CI 0.11–2.63; AP: 0.43, 95% CI 0.17–0.69; and SI:2.68, 95% CI 1.10–6.48). The interactive effect between HGI and abdominal obesity was also found to be significant, as estimated by the value of RERI (1.04, 95% CI 0.24–1.85), AP (0.33, 95% CI 0.11–0.56) and SI (1.96, 95% CI 1.01–3.79). However, in the analysis of the interaction between HGI and general obesity, only the AP value (0.28, 95% CI 0.01–0.54) was observed to be significant. Conclusion High HGI was independently associated with the risk of hypertension. Moreover, HGI significantly shared interactions with obesity and family history of hypertension that influenced the risk of hypertension.
Collapse
Affiliation(s)
- Jing Mi
- School of Public Health, Bengbu Medical College, 2600 Donghai road, Bengbu, 233000, Anhui Province, China
| | - Jian Song
- School of Public Health, Bengbu Medical College, 2600 Donghai road, Bengbu, 233000, Anhui Province, China
| | - Yingying Zhao
- Bengbu Health Board, 568 Nanhu road, Bengbu, 233000, Anhui Province, China
| | - Xuesen Wu
- School of Public Health, Bengbu Medical College, 2600 Donghai road, Bengbu, 233000, Anhui Province, China.
| |
Collapse
|
79
|
Aragonès G, Rowan S, G Francisco S, Yang W, Weinberg J, Taylor A, Bejarano E. Glyoxalase System as a Therapeutic Target against Diabetic Retinopathy. Antioxidants (Basel) 2020; 9:antiox9111062. [PMID: 33143048 PMCID: PMC7692619 DOI: 10.3390/antiox9111062] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Hyperglycemia, a defining characteristic of diabetes, combined with oxidative stress, results in the formation of advanced glycation end products (AGEs). AGEs are toxic compounds that have adverse effects on many tissues including the retina and lens. AGEs promote the formation of reactive oxygen species (ROS), which, in turn, boost the production of AGEs, resulting in positive feedback loops, a vicious cycle that compromises tissue fitness. Oxidative stress and the accumulation of AGEs are etiologically associated with the pathogenesis of multiple diseases including diabetic retinopathy (DR). DR is a devastating microvascular complication of diabetes mellitus and the leading cause of blindness in working-age adults. The onset and development of DR is multifactorial. Lowering AGEs accumulation may represent a potential therapeutic approach to slow this sight-threatening diabetic complication. To set DR in a physiological context, in this review we first describe relations between oxidative stress, formation of AGEs, and aging in several tissues of the eye, each of which is associated with a major age-related eye pathology. We summarize mechanisms of AGEs generation and anti-AGEs detoxifying systems. We specifically feature the potential of the glyoxalase system in the retina in the prevention of AGEs-associated damage linked to DR. We provide a comparative analysis of glyoxalase activity in different tissues from wild-type mice, supporting a major role for the glyoxalase system in the detoxification of AGEs in the retina, and present the manipulation of this system as a therapeutic strategy to prevent the onset of DR.
Collapse
Affiliation(s)
- Gemma Aragonès
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
| | - Sheldon Rowan
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02155, USA
- Friedman School of Nutrition and Science Policy, Tufts University, Boston, MA 02155, USA
| | - Sarah G Francisco
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
| | - Wenxin Yang
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
| | - Jasper Weinberg
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
| | - Allen Taylor
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02155, USA
- Friedman School of Nutrition and Science Policy, Tufts University, Boston, MA 02155, USA
- Correspondence: (A.T.); (E.B.); Tel.: +617-556-3156 (A.T.)
| | - Eloy Bejarano
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02155, USA; (G.A.); (S.R.); (S.G.F.); (W.Y.); (J.W.)
- Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Valencia, Spain
- Correspondence: (A.T.); (E.B.); Tel.: +617-556-3156 (A.T.)
| |
Collapse
|
80
|
Sisay M, Edessa D, Ali T, Mekuria AN, Gebrie A. The relationship between advanced glycation end products and gestational diabetes: A systematic review and meta-analysis. PLoS One 2020; 15:e0240382. [PMID: 33085688 PMCID: PMC7577486 DOI: 10.1371/journal.pone.0240382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Gestational Diabetes Mellitus (GDM) is a condition in which women without history of diabetes experience hyperglycemia during pregnancy, especially at the second and third trimesters. In women who have had GDM, an elevated body mass index (BMI) may have a substantial impact for persistent hyperglycemia in their lives after gestation. Beyond hyperglycemia, increased local oxidative stress directly promotes the formation of Advanced Glycation End-products (AGEs). Hence, this systematic review and meta-analysis was aimed to determine the relationship between the level of AGEs and/or related metabolic biomarkers with GDM. METHODS Literature search was carried out through visiting electronic databases, indexing services, and directories including PubMed/MEDLINE (Ovid®), EMBASE (Ovid®), google scholar and WorldCat to retrieve studies without time limit. Following screening and eligibility evaluation, relevant data were extracted from included studies and analyzed using Rev-Man 5.3 and STATA 15.0. Inverse variance method with random effects pooling model was used for the analysis of outcome measures at 95% confidence interval. Hedge's adjusted g statistics was applied to calculate the standardized mean difference (SMD) to consider the small sample bias. Besides, meta-regression, meta-influence, and publication bias analyses were conducted. The protocol has been registered on PROSPERO with ID: CRD42020173867. RESULTS A total of 16 original studies were included for the systematic review and meta-analysis. Compared with women with pregnant controls, the level of AGE was significantly higher in women with GDM (SMD [95% CI] = 2.26 [1.50‒3.02], Z = 5.83, P < 0.00001; I2 = 97%, P< 0.0001). The BMI was also significantly higher in women with GDM (SMD [95% CI] = 0.97 [0.33‒1.62], Z = 2.98, P = 0.003) compared to controls. Regarding specific and related metabolic biomarkers, there was higher level of HOMA-IR (SMD [95% CI] = 0.39 [0.22-0.55], Z = 4.65, P < 0.0001, after sensitivity analysis) and HbA1c (SMD [95% CI] = 0.58 [0.03‒1.12], Z = 2.07, P = 0.04, after sensitivity analysis) in gestational diabetic women. Subgroup analyses indicated that studies conducted in Asia and Europe, at third trimester of pregnancy and blood/plasma AGE samples showed a significant difference in AGE level among women with GDM compared to pregnant controls. What is more, meta-regression with the sample size (regression coefficient (Q) = -0.0092, P = 0.207) and year of publication (Q = 0.0035, P = 0.984) suggested that the covariates had no significant effect on the heterogeneity. CONCLUSION The study indicated that there was a strong relationship between AGE and GDM. Besides, the BMI and other specific biomarkers showed a significant difference between the two groups indicating the high risk of developing long-standing type 2 diabetes and its complications in gestational diabetic women. Early detection of these biomarkers may play a pivotal role in controlling postpartum diabetic complications.
Collapse
Affiliation(s)
- Mekonnen Sisay
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Dumessa Edessa
- Department of Clinical Pharmacy, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Tilahun Ali
- Department of Psychiatry, School of Nursing and Midwifery, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Abraham Nigussie Mekuria
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Alemu Gebrie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
81
|
Cordova R, Knaze V, Viallon V, Rust P, Schalkwijk CG, Weiderpass E, Wagner KH, Mayen-Chacon AL, Aglago EK, Dahm CC, Overvad K, Tjønneland A, Halkjær J, Mancini FR, Boutron-Ruault MC, Fagherazzi G, Katzke V, Kühn T, Schulze MB, Boeing H, Trichopoulou A, Karakatsani A, Thriskos P, Masala G, Krogh V, Panico S, Tumino R, Ricceri F, Spijkerman A, Boer J, Skeie G, Rylander C, Borch KB, Quirós JR, Agudo A, Redondo-Sánchez D, Amiano P, Gómez-Gómez JH, Barricarte A, Ramne S, Sonestedt E, Johansson I, Esberg A, Tong T, Aune D, Tsilidis KK, Gunter MJ, Jenab M, Freisling H. Dietary intake of advanced glycation end products (AGEs) and changes in body weight in European adults. Eur J Nutr 2020; 59:2893-2904. [PMID: 31701336 DOI: 10.1007/s00394-019-02129-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/23/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Advanced glycation end products (AGEs) can be formed in foods by the reaction of reducing sugars with proteins, and have been shown to induce insulin resistance and obesity in experimental studies. We examined the association between dietary AGEs intake and changes in body weight in adults over an average of 5 years of follow-up. METHODS A total of 255,170 participants aged 25-70 years were recruited in ten European countries (1992-2000) in the PANACEA study (Physical Activity, Nutrition, Alcohol, Cessation of smoking, Eating out of home in relation to Anthropometry), a sub-cohort of the EPIC (European Prospective Investigation into Cancer and Nutrition). Body weight was measured at recruitment and self-reported between 2 and 11 years later depending on the study center. A reference database for AGEs was used containing UPLC-MS/MS-measured Nε-(carboxymethyl)-lysine (CML), Nε-(1-carboxyethyl)-lysine (CEL), and Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1) in 200 common European foods. This reference database was matched to foods and decomposed recipes obtained from country-specific validated dietary questionnaires in EPIC and intake levels of CEL, CML, and MG-H1 were estimated. Associations between dietary AGEs intake and body weight change were estimated separately for each of the three AGEs using multilevel mixed linear regression models with center as random effect and dietary AGEs intake and relevant confounders as fixed effects. RESULTS A one-SD increment in CEL intake was associated with 0.111 kg (95% CI 0.087-0.135) additional weight gain over 5 years. The corresponding additional weight gain for CML and MG-H1 was 0.065 kg (0.041-0.089) and 0.034 kg (0.012, 0.057), respectively. The top six food groups contributing to AGEs intake, with varying proportions across the AGEs, were cereals/cereal products, meat/processed meat, cakes/biscuits, dairy, sugar and confectionary, and fish/shellfish. CONCLUSION In this study of European adults, higher intakes of AGEs were associated with marginally greater weight gain over an average of 5 years of follow-up.
Collapse
Affiliation(s)
- R Cordova
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - V Knaze
- Section of Early Detection and Prevention, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - V Viallon
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - P Rust
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - C G Schalkwijk
- Department of Internal Medicine, Laboratory of Metabolism and Vascular Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - E Weiderpass
- International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - K-H Wagner
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - A-L Mayen-Chacon
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - E K Aglago
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - C C Dahm
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - K Overvad
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - A Tjønneland
- Danish Cancer Society Research Center Copenhagen, Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - J Halkjær
- Danish Cancer Society Research Center Copenhagen, Copenhagen, Denmark
| | - F R Mancini
- CESP, Fac. de médecine, Univ. Paris-Sud, Fac. de médecine-UVSQ-INSERM, Université Paris-Saclay, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
| | - M-C Boutron-Ruault
- CESP, Fac. de médecine, Univ. Paris-Sud, Fac. de médecine-UVSQ-INSERM, Université Paris-Saclay, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
| | - G Fagherazzi
- CESP, Fac. de médecine, Univ. Paris-Sud, Fac. de médecine-UVSQ-INSERM, Université Paris-Saclay, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
| | - V Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutrition Science, University of Potsdam, Nuthetal, Germany
| | - H Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | | | - A Karakatsani
- Hellenic Health Foundation, Athens, Greece
- 2nd Pulmonary Medicine Department, School of Medicine, National and Kapodistrian University of Athens, ATTIKON University Hospital, Haidari, Greece
| | - P Thriskos
- Hellenic Health Foundation, Athens, Greece
| | - G Masala
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network-ISPRO, Florence, Italy
| | - V Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Panico
- Dipartimento di Medicina Clinica E Chirurgia, Federico II University, Naples, Italy
| | - R Tumino
- Cancer Registry and Histopathology Unit, Azienda Sanitaria Provinciale (ASP) Ragusa, Ragusa, Italy
| | - F Ricceri
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Unit of Epidemiology, Regional Health Service ASL TO3, Turin, TO, Italy
| | - A Spijkerman
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - J Boer
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - G Skeie
- Department of Community Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - C Rylander
- Department of Community Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - K B Borch
- Department of Community Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - J R Quirós
- Public Health Directorate, Asturias, Spain
| | - A Agudo
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - D Redondo-Sánchez
- Andalusian School of Public Health. Biomedical Research Institute ibs.GRANADA, University of Granada, Granada, Spain
- CIBER of Epidemiology and Public Health, Madrid, Spain
| | - P Amiano
- CIBER of Epidemiology and Public Health, Madrid, Spain
- Public Health Division of Gipuzkoa, BioDonostia Research Institute, San Sebastian, Spain
| | - J-H Gómez-Gómez
- Department of Epidemiology and Murcia Regional Health Council, Universidad de Murcia, Murcia, Spain
| | - A Barricarte
- CIBER of Epidemiology and Public Health, Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - S Ramne
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - E Sonestedt
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - I Johansson
- Department of Odontology, Umeå University, Umeå, Sweden
| | - A Esberg
- Department of Odontology, Umeå University, Umeå, Sweden
| | - T Tong
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - D Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - K K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine University Campus Ioannina, Ioannina, Greece
| | - M J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - M Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Heinz Freisling
- International Agency for Research on Cancer (IARC-WHO), 150 cours Albert Thomas, CEDEX 08, 69372, Lyon, France.
| |
Collapse
|
82
|
Wu Q, Coumoul X, Grandjean P, Barouki R, Audouze K. Endocrine disrupting chemicals and COVID-19 relationships: a computational systems biology approach. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.07.10.20150714. [PMID: 32699854 PMCID: PMC7373141 DOI: 10.1101/2020.07.10.20150714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Patients at high risk of severe forms of COVID-19 frequently suffer from chronic diseases, but other risk factors may also play a role. Environmental stressors, such as endocrine disrupting chemicals (EDCs), can contribute to certain chronic diseases and might aggravate the course of COVID-19. Objectives To explore putative links between EDCs and COVID-19 severity, an integrative systems biology approach was constructed and applied. Methods As a first step, relevant data sets were compiled from major data sources. Biological associations of major EDCs to proteins were extracted from the CompTox database. Associations between proteins and diseases known as important COVID-19 comorbidities were obtained from the GeneCards and DisGeNET databases. Based on these data, we developed a tripartite network (EDCs-proteins-diseases) and used it to identify proteins overlapping between the EDCs and the diseases. Signaling pathways for common proteins were then investigated by over-representation analysis. Results We found several statistically significant pathways that may be dysregulated by EDCs and that may also be involved in COVID-19 severity. The Th17 and the AGE/RAGE signaling pathways were particularly promising. Conclusions Pathways were identified as possible targets of EDCs and as contributors to COVID-19 severity, thereby highlighting possible links between exposure to environmental chemicals and disease development. This study also documents the application of computational systems biology methods as a relevant approach to increase the understanding of molecular mechanisms linking EDCs and human diseases, thereby contributing to toxicology prediction.
Collapse
Affiliation(s)
- Qier Wu
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Xavier Coumoul
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Philippe Grandjean
- Harvard T.H.Chan School of Public Health, Boston, MA 02215, USA
- University of Southern Denmark, 5000 Odense C, Denmark
| | - Robert Barouki
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Karine Audouze
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| |
Collapse
|
83
|
Zhang Q, Wang Y, Fu L. Dietary advanced glycation end‐products: Perspectives linking food processing with health implications. Compr Rev Food Sci Food Saf 2020; 19:2559-2587. [DOI: 10.1111/1541-4337.12593] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/07/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Qiaozhi Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and BiotechnologyZhejiang Gongshang University Hangzhou P.R. China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and BiotechnologyZhejiang Gongshang University Hangzhou P.R. China
| | - Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and BiotechnologyZhejiang Gongshang University Hangzhou P.R. China
| |
Collapse
|
84
|
Gutierrez-Mariscal FM, Cardelo MP, de la Cruz S, Alcala-Diaz JF, Roncero-Ramos I, Guler I, Vals-Delgado C, López-Moreno A, Luque RM, Delgado-Lista J, Perez-Martinez P, Yubero-Serrano EM, Lopez-Miranda J. Reduction in Circulating Advanced Glycation End Products by Mediterranean Diet Is Associated with Increased Likelihood of Type 2 Diabetes Remission in Patients with Coronary Heart Disease: From the Cordioprev Study. Mol Nutr Food Res 2020; 65:e1901290. [PMID: 32529753 DOI: 10.1002/mnfr.201901290] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/01/2020] [Indexed: 01/26/2023]
Abstract
SCOPE It is hypothesized that decreased advanced glycation end products (AGEs) levels could affect type 2 diabetes mellitus (T2DM) remission in newly diagnosed patients through the consumption of two healthy diets. METHODS AND RESULTS Patients from CORDIOPREV study, all with previous cardiovascular events, with T2DM at the beginning of the study are included. Patients are randomized to a Mediterranean or a low-fat diet for five years. No different diabetes remission rates are found among diets. Serum methylglioxal (MG) and carboximethyllysine (CML), levels dietary AGE, as well as gene expression of AGER1 and RAGE are measured. Serum MG decreases only after the consumption of the Mediterranean diet. Moreover, a COX regression analysis shows that each SD decrease in the MG, occurring after the Mediterranean diet, increases the probability of T2DM remission with HR:2.56(1.02-6.25) and p = 0.046 and each SD increase in disposition index at baseline increases the probability of remission with HR:1.94(1.32-2.87) and p = 0.001. CONCLUSIONS It is demonstrated that the reduction of serum AGEs levels and the modulation of its metabolism, occurring after the consumption of a Mediterranean diet, might be involved in the molecular mechanism underlying the T2DM remission of newly diagnosed patients with coronary heart disease.
Collapse
Affiliation(s)
- Francisco M Gutierrez-Mariscal
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Magdalena P Cardelo
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Silvia de la Cruz
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Juan F Alcala-Diaz
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Irene Roncero-Ramos
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Ipek Guler
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,Department of Innovation and Methodology, IMIBIC, Córdoba, 14004, Spain
| | - Cristina Vals-Delgado
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Alejandro López-Moreno
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Raul M Luque
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, 14071, Spain
| | - Javier Delgado-Lista
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Pablo Perez-Martinez
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Elena M Yubero-Serrano
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| | - Jose Lopez-Miranda
- Maimonines Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, 14004, Spain.,Hospital Universitario Reina Sofia (HURS), Córdoba, 14004, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, 28029, Spain.,Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, University of Córdoba, Córdoba, 14004, Spain
| |
Collapse
|
85
|
Griffith RJ, Alsweiler J, Moore AE, Brown S, Middleton P, Shepherd E, Crowther CA. Interventions to prevent women from developing gestational diabetes mellitus: an overview of Cochrane Reviews. Cochrane Database Syst Rev 2020; 6:CD012394. [PMID: 32526091 PMCID: PMC7388385 DOI: 10.1002/14651858.cd012394.pub3] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The prevalence of gestational diabetes mellitus (GDM) is increasing, with approximately 15% of pregnant women affected worldwide, varying by country, ethnicity and diagnostic thresholds. There are associated short- and long-term health risks for women and their babies. OBJECTIVES We aimed to summarise the evidence from Cochrane systematic reviews on the effects of interventions for preventing GDM. METHODS We searched the Cochrane Database of Systematic Reviews (6 August 2019) with key words 'gestational diabetes' OR 'GDM' to identify reviews pre-specifying GDM as an outcome. We included reviews of interventions in women who were pregnant or planning a pregnancy, irrespective of their GDM risk status. Two overview authors independently assessed eligibility, extracted data and assessed quality of evidence using ROBIS and GRADE tools. We assigned interventions to categories with graphic icons to classify the effectiveness of interventions as: clear evidence of benefit or harm (GRADE moderate- or high-quality evidence with a confidence interval (CI) that did not cross the line of no effect); clear evidence of no effect or equivalence (GRADE moderate- or high-quality evidence with a narrow CI crossing the line of no effect); possible benefit or harm (low-quality evidence with a CI that did not cross the line of no effect or GRADE moderate- or high-quality evidence with a wide CI); or unknown benefit or harm (GRADE low-quality evidence with a wide CI or very low-quality evidence). MAIN RESULTS We included 11 Cochrane Reviews (71 trials, 23,154 women) with data on GDM. Nine additional reviews pre-specified GDM as an outcome, but did not identify GDM data in included trials. Ten of the 11 reviews were judged to be at low risk of bias and one review at unclear risk of bias. Interventions assessed included diet, exercise, a combination of diet and exercise, dietary supplements, pharmaceuticals, and management of other health problems in pregnancy. The quality of evidence ranged from high to very low. Diet Unknown benefit or harm: there was unknown benefit or harm of dietary advice versus standard care, on the risk of GDM: risk ratio (RR) 0.60, 95% CI 0.35 to 1.04; 5 trials; 1279 women; very low-quality evidence. There was unknown benefit or harm of a low glycaemic index diet versus a moderate-high glycaemic index diet on the risk of GDM: RR 0.91, 95% CI 0.63 to 1.31; 4 trials; 912 women; low-quality evidence. Exercise Unknown benefit or harm: there was unknown benefit or harm for exercise interventions versus standard antenatal care on the risk of GDM: RR 1.10, 95% CI 0.66 to 1.84; 3 trials; 826 women; low-quality evidence. Diet and exercise combined Possible benefit: combined diet and exercise interventions during pregnancy versus standard care possibly reduced the risk of GDM: RR 0.85, 95% CI 0.71 to 1.01; 19 trials; 6633 women; moderate-quality evidence. Dietary supplements Clear evidence of no effect: omega-3 fatty acid supplementation versus none in pregnancy had no effect on the risk of GDM: RR 1.02, 95% CI 0.83 to 1.26; 12 trials; 5235 women; high-quality evidence. Possible benefit: myo-inositol supplementation during pregnancy versus control possibly reduced the risk of GDM: RR 0.43, 95% CI 0.29 to 0.64; 3 trials; 502 women; low-quality evidence. Possible benefit: vitamin D supplementation versus placebo or control in pregnancy possibly reduced the risk of GDM: RR 0.51, 95% CI 0.27 to 0.97; 4 trials; 446 women; low-quality evidence. Unknown benefit or harm: there was unknown benefit or harm of probiotic with dietary intervention versus placebo with dietary intervention (RR 0.37, 95% CI 0.15 to 0.89; 1 trial; 114 women; very low-quality evidence), or probiotic with dietary intervention versus control (RR 0.38, 95% CI 0.16 to 0.92; 1 trial; 111 women; very low-quality evidence) on the risk of GDM. There was unknown benefit or harm of vitamin D + calcium supplementation versus placebo (RR 0.33, 95% CI 0.01 to 7.84; 1 trial; 54 women; very low-quality evidence) or vitamin D + calcium + other minerals versus calcium + other minerals (RR 0.42, 95% CI 0.10 to 1.73; 1 trial; 1298 women; very low-quality evidence) on the risk of GDM. Pharmaceutical Possible benefit: metformin versus placebo given to obese pregnant women possibly reduced the risk of GDM: RR 0.85, 95% CI 0.61 to 1.19; 3 trials; 892 women; moderate-quality evidence. Unknown benefit or harm:eight small trials with low- to very low-quality evidence showed unknown benefit or harm for heparin, aspirin, leukocyte immunisation or IgG given to women with a previous stillbirth on the risk of GDM. Management of other health issues Clear evidence of no effect: universal versus risk based screening of pregnant women for thyroid dysfunction had no effect on the risk of GDM: RR 0.93, 95% CI 0.70 to 1.25; 1 trial; 4516 women; moderate-quality evidence. Unknown benefit or harm: there was unknown benefit or harm of using fractional exhaled nitrogen oxide versus a clinical algorithm to adjust asthma therapy on the risk of GDM: RR 0.74, 95% CI 0.31 to 1.77; 1 trial; 210 women; low-quality evidence. There was unknown benefit or harm of pharmacist led multidisciplinary approach to management of maternal asthma versus standard care on the risk of GDM: RR 5.00, 95% CI 0.25 to 99.82; 1 trial; 58 women; low-quality evidence. AUTHORS' CONCLUSIONS No interventions to prevent GDM in 11 systematic reviews were of clear benefit or harm. A combination of exercise and diet, supplementation with myo-inositol, supplementation with vitamin D and metformin were of possible benefit in reducing the risk of GDM, but further high-quality evidence is needed. Omega-3-fatty acid supplementation and universal screening for thyroid dysfunction did not alter the risk of GDM. There was insufficient high-quality evidence to establish the effect on the risk of GDM of diet or exercise alone, probiotics, vitamin D with calcium or other vitamins and minerals, interventions in pregnancy after a previous stillbirth, and different asthma management strategies in pregnancy. There is a lack of trials investigating the effect of interventions prior to or between pregnancies on risk of GDM.
Collapse
Affiliation(s)
- Rebecca J Griffith
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, New Zealand
| | - Jane Alsweiler
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, New Zealand
| | - Abigail E Moore
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Stephen Brown
- School of Interprofessional Health Studies, Auckland University of Technology, Auckland, New Zealand
| | - Philippa Middleton
- Healthy Mothers, Babies and Children, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Emily Shepherd
- Robinson Research Institute, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | | |
Collapse
|
86
|
Tyagi A, Mirita C, Taher N, Shah I, Moeller E, Tyagi A, Chong T, Pugazhenthi S. Metabolic syndrome exacerbates amyloid pathology in a comorbid Alzheimer's mouse model. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165849. [PMID: 32485218 DOI: 10.1016/j.bbadis.2020.165849] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) often coexists with other aging-associated diseases including obesity, diabetes, hypertension, and cardiovascular diseases. The early stage of these comorbidities is known as metabolic syndrome (MetS) which is highly prevalent in mid-life. An important cause of MetS is the deficiency of SIRT3, a mitochondrial deacetylase which enhances the functions of critical mitochondrial proteins, including metabolic enzymes, by deacetylation. Deletion of Sirt3 gene has been reported to result in the acceleration of MetS. In a recently published study, we demonstrated in the brain of Sirt3-/- mice, downregulation of metabolic enzymes, insulin resistance and elevation of inflammatory markers including microglial proliferation. These findings suggested a novel pathway that could link SIRT3 deficiency to neuroinflammation, an important cause of Alzheimer's pathogenesis. Therefore, we hypothesized that MetS and amyloid pathology may interact through converging pathways of insulin resistance and neuroinflammation in comorbid AD. To investigate these interactions, we crossed Sirt3-/- mice with APP/PS1 mice and successfully generated APP/PS1/Sirt3-/- mice with amyloid pathology and MetS. In these comorbid AD mice, we observed exacerbation of insulin resistance, glucose intolerance, amyloid plaque deposition, markers of neuroinflammation, including elevated expression of IL-1β, TNF-α and Cox-2 at 8 months of age. There was also increased microglial proliferation and activation. Our observations suggest a novel mechanism by which MetS may interact with amyloid pathology during the cellular phase of AD. Therapeutic targeting of SIRT3 in AD with comorbidities may produce beneficial effects.
Collapse
Affiliation(s)
- Alpna Tyagi
- Rocky Mountain Regional VA Medical Center, USA; Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Iman Shah
- Rocky Mountain Regional VA Medical Center, USA
| | | | - Anit Tyagi
- Rocky Mountain Regional VA Medical Center, USA; Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | | | - Subbiah Pugazhenthi
- Rocky Mountain Regional VA Medical Center, USA; Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
87
|
Sergi D, Boulestin H, Campbell FM, Williams LM. The Role of Dietary Advanced Glycation End Products in Metabolic Dysfunction. Mol Nutr Food Res 2020; 65:e1900934. [PMID: 32246887 DOI: 10.1002/mnfr.201900934] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/09/2020] [Indexed: 12/13/2022]
Abstract
Advanced glycation end products (AGEs) are a heterogeneous group of molecules produced, non-enzymatically, from the interaction between reducing sugars and the free amino groups of proteins, nucleic acids, and lipids. AGEs are formed as a normal consequence of metabolism but can also be absorbed from the diet. They have been widely implicated in the complications of diabetes affecting cardiovascular health, the nervous system, eyes, and kidneys. Increased levels of AGEs are also detrimental to metabolic health and may contribute to the metabolic abnormalities induced by the Western diet, which is high in processed foods and represents a significant source of AGEs. While increased AGE levels are a consequence of diabetic hyperglycaemia, AGEs themselves activate signaling pathways, which compromise insulin signaling and pancreatic β-cell function, thus, contributing to the development of type 2 diabetes mellitus (T2DM). Furthermore, AGEs may also contribute to the obesogenic effects of the Western diet by promoting hypothalamic inflammation and disrupting the central control of energy balance. Here, the role of dietary AGEs in metabolic dysfunction is reviewed with a focus on the mechanisms underpinning their detrimental role in insulin resistance, pancreatic β-cell dysfunction, hypothalamic control of energy balance, and the pathogenesis of T2DM and obesity.
Collapse
Affiliation(s)
- Domenico Sergi
- Nutrition and Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, 5000, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Hakim Boulestin
- Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Fiona M Campbell
- Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Lynda M Williams
- Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
88
|
Velayoudom-Cephise FL, Cano-Sanchez M, Bercion S, Tessier F, Yu Y, Boulanger E, Neviere R. Receptor for advanced glycation end products modulates oxidative stress and mitochondrial function in the soleus muscle of mice fed a high-fat diet. Appl Physiol Nutr Metab 2020; 45:1107-1117. [PMID: 32289236 DOI: 10.1139/apnm-2019-0936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulation of advanced glycation end products (AGEs) and activation of the receptor for AGEs (RAGE) are implicated in the progression of pathologies associated with aging, chronic inflammation, diabetes, and cellular stress. RAGE activation is also implicated in cardiovascular complications of type 2 diabetes, such as nephropathy, retinopathy, accelerated vascular diseases, and cardiomyopathy. Studies investigating the effects of AGE/RAGE axis activation on skeletal muscle oxidative stress and metabolism are more limited. We tested whether a high-fat diet (HFD) would alter circulating AGE concentration, skeletal muscle AGE accumulation, and oxidative stress in wild-type and RAGE-deficient mice. The physiological significance of AGE/RAGE axis activation in HFD-fed mice was evaluated in terms of exercise tolerance and mitochondrial respiratory chain complex activity. HFD elicited adiposity, abnormal fat distribution, and oral glucose intolerance. HFD also induced accumulation of Nε-carboxymethyl-l-lysine, increased protein carbonyl levels, and impaired respiratory chain complex activity in soleus muscle. Ablation of RAGE had no effects on weight gain and oral glucose tolerance in HFD-fed mice. Peak aerobic capacity and mitochondrial cytochrome-c oxidase activity were restored in HFD-fed RAGE-/- mice. We concluded that RAGE signaling plays an important role in skeletal muscle homeostasis of mice under metabolic stress. Novelty HFD in mice induces accumulation of AGEs, oxidative stress, and mitochondrial dysfunction in the soleus muscle. RAGE, the multi-ligand receptor for AGEs, modulates oxidative stress and mitochondrial electron transport chain function in the soleus muscle of HFD-fed mice.
Collapse
Affiliation(s)
- Fritz Line Velayoudom-Cephise
- University Hospital CHU of Guadeloupe, Pointe à Pitre, 97110, France.,EA7525, University of the French West Indies, Fort de France, 97159, France
| | - Mariola Cano-Sanchez
- EA7525, University of the French West Indies, Fort de France, 97159, France.,University Hospital CHU of Martinique, Fort de France, 97200, France
| | - Sylvie Bercion
- EA7525, University of the French West Indies, Fort de France, 97159, France.,Department of Chemistry, Faculty of Natural Sciences, Pointe a Pitre, 97110, France
| | - Frédéric Tessier
- INSERM U995, LIRIC Team "Glycation: from inflammation to aging", Lille University, Lille, 59000, France
| | - Yichi Yu
- INSERM U995, LIRIC Team "Glycation: from inflammation to aging", Lille University, Lille, 59000, France.,School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Eric Boulanger
- INSERM U995, LIRIC Team "Glycation: from inflammation to aging", Lille University, Lille, 59000, France
| | - Remi Neviere
- EA7525, University of the French West Indies, Fort de France, 97159, France.,University Hospital CHU of Martinique, Fort de France, 97200, France
| |
Collapse
|
89
|
Yubero-Serrano EM, Pérez-Martínez P. Advanced Glycation End Products and Their Involvement in Cardiovascular Disease. Angiology 2020; 71:698-700. [DOI: 10.1177/0003319720916301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Elena M. Yubero-Serrano
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba, Reina Sofia University Hospital, University of Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Pablo Pérez-Martínez
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba, Reina Sofia University Hospital, University of Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
90
|
Zhao Z, Li M, Li C, Wang T, Xu Y, Zhan Z, Dong W, Shen Z, Xu M, Lu J, Chen Y, Lai S, Fan W, Bi Y, Wang W, Ning G. Dietary preferences and diabetic risk in China: A large-scale nationwide Internet data-based study. J Diabetes 2020; 12:270-278. [PMID: 31290214 DOI: 10.1111/1753-0407.12967] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/21/2019] [Accepted: 07/05/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Unhealthy diet is one of the important risk factors of diabetes, which is one of the major public health problems in China. The Internet tools provide large-scale passively collected data that show people's dietary preferences and their relationship with diabetes risk. METHODS 212 341 708 individuals' dietary preference labels were created based on Internet data from online search and shopping software. Metabolic data obtained from the 2010 China Noncommunicable Disease Surveillance, which had 98 658 participants, was used to estimate the relation between dietary preferences geographical distribution and diabetes risk. RESULTS Chinese dietary preferences had different geographical distribution, which is related to the local climate and consumption level. Fried food preference proportion distribution was significantly positively correlated with diabetes prevalence, hypertension prevalence and body mass index (BMI). Similarly, grilled food preference proportion distribution had significantly positive correlation with the prevalence of diabetes and hypertension. In contrast, spicy food preference proportion distribution was negatively correlated with diabetes prevalence. Sweet food preference proportion distribution was positively related to diabetes prevalence. Using dietary preferences data to predict regional prevalence of diabetes, hypertension and BMI, the average values of error (95% CI) between the three paired predicted and observed values were 9.8% (6.9%-12.7%), 7.5% (5.0%-10.0%) and 1.6% (1.2%-2.0%), respectively. CONCLUSIONS Fried food, grilled food, and sweet food preferences were positively related to diabetes risk whereas spicy food preference was negatively correlated with diabetes risk. Dietary preferences based on passively collected Internet data could be used to predict regional prevalence of diabetes, hypertension, and BMI and showed good value for public health monitoring.
Collapse
Affiliation(s)
- Zhiyun Zhao
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Mian Li
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Chao Li
- Beijing Baidu Netcom Science & Technology Co., Ltd., Beijing, China
| | - Tiange Wang
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Zhizheng Zhan
- Beijing Baidu Netcom Science & Technology Co., Ltd., Beijing, China
| | - Weishan Dong
- Beijing Baidu Netcom Science & Technology Co., Ltd., Beijing, China
| | - Zhiyong Shen
- Beijing Baidu Netcom Science & Technology Co., Ltd., Beijing, China
| | - Min Xu
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Shenghan Lai
- Department of Pathology, Radiology, Epidemiology & Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Wei Fan
- Beijing Baidu Netcom Science & Technology Co., Ltd., Beijing, China
| | - Yufang Bi
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- State Key Laboratory of Medical Genomics, and National Clinical Research Center for Metabolic Diseases, Rui-Jin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| |
Collapse
|
91
|
Wang X, Liu J, Yang Y, Zhang X. An update on the potential role of advanced glycation end products in glycolipid metabolism. Life Sci 2020; 245:117344. [DOI: 10.1016/j.lfs.2020.117344] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
|
92
|
Cepas V, Collino M, Mayo JC, Sainz RM. Redox Signaling and Advanced Glycation Endproducts (AGEs) in Diet-Related Diseases. Antioxidants (Basel) 2020; 9:antiox9020142. [PMID: 32041293 PMCID: PMC7070562 DOI: 10.3390/antiox9020142] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/19/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Diets are currently characterized by elevated sugar intake, mainly due to the increased consumption of processed sweetened foods and drinks during the last 40 years. Diet is the main source of advanced glycation endproducts (AGEs). These are toxic compounds formed during the Maillard reaction, which takes place both in vivo, in tissues and fluids under physiological conditions, favored by sugar intake, and ex vivo during food preparation such as baking, cooking, frying or storage. Protein glycation occurs slowly and continuously through life, driving AGE accumulation in tissues during aging. For this reason, AGEs have been proposed as a risk factor in the pathogenesis of diet-related diseases such as diabetes, insulin resistance, cardiovascular diseases, kidney injury, and age-related and neurodegenerative diseases. AGEs are associated with an increase in oxidative stress since they mediate the production of reactive oxygen species (ROS), increasing the intracellular levels of hydrogen peroxide (H2O2), superoxide (O2−), and nitric oxide (NO). The interaction of AGEs with the receptor for AGEs (RAGE) enhances oxidative stress through ROS production by NADPH oxidases inside the mitochondria. This affects mitochondrial function and ultimately influences cell metabolism under various pathological conditions. This short review will summarize all evidence that relates AGEs and ROS production, their relationship with diet-related diseases, as well as the latest research about the use of natural compounds with antioxidant properties to prevent the harmful effects of AGEs on health.
Collapse
Affiliation(s)
- Vanesa Cepas
- Departamento de Morfologia y Biologia Celular, Redox Biology Group, Universidad de Oviedo, 33403 Oviedo, Spain;
- Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, 33403 Oviedo, Spain
| | - Massimo Collino
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy;
| | - Juan C. Mayo
- Departamento de Morfologia y Biologia Celular, Redox Biology Group, Universidad de Oviedo, 33403 Oviedo, Spain;
- Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, 33403 Oviedo, Spain
- Correspondence: (J.C.M.); (R.M.S.); Tel.: +34-985-10-2730 (J.C.M.); +34-985-10-3610 (R.M.S.)
| | - Rosa M. Sainz
- Departamento de Morfologia y Biologia Celular, Redox Biology Group, Universidad de Oviedo, 33403 Oviedo, Spain;
- Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, 33403 Oviedo, Spain
- Correspondence: (J.C.M.); (R.M.S.); Tel.: +34-985-10-2730 (J.C.M.); +34-985-10-3610 (R.M.S.)
| |
Collapse
|
93
|
Ruiz HH, Ramasamy R, Schmidt AM. Advanced Glycation End Products: Building on the Concept of the "Common Soil" in Metabolic Disease. Endocrinology 2020; 161:bqz006. [PMID: 31638645 PMCID: PMC7188081 DOI: 10.1210/endocr/bqz006] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 10/01/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
The role of advanced glycation end products (AGEs) in promoting and/or exacerbating metabolic dysregulation is being increasingly recognized. AGEs are formed when reducing sugars nonenzymatically bind to proteins or lipids, a process that is enhanced by hyperglycemic and hyperlipidemic environments characteristic of numerous metabolic disorders including obesity, diabetes, and its complications. In this mini-review, we put forth the notion that AGEs span the spectrum from cause to consequence of insulin resistance and diabetes, and represent a "common soil" underlying the pathophysiology of these metabolic disorders. Collectively, the surveyed literature suggests that AGEs, both those that form endogenously as well as exogenous AGEs derived from environmental factors such as pollution, smoking, and "Western"-style diets, contribute to the pathogenesis of obesity and diabetes. Specifically, AGE accumulation in key metabolically relevant organs induces insulin resistance, inflammation, and oxidative stress, which in turn provide substrates for excess AGE formation, thus creating a feed-forward-fueled pathological loop mediating metabolic dysfunction.
Collapse
Affiliation(s)
- Henry H Ruiz
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU School of Medicine, New York, NY, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU School of Medicine, New York, NY, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
94
|
Schalkwijk CG, Stehouwer CDA. Methylglyoxal, a Highly Reactive Dicarbonyl Compound, in Diabetes, Its Vascular Complications, and Other Age-Related Diseases. Physiol Rev 2020; 100:407-461. [DOI: 10.1152/physrev.00001.2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The formation and accumulation of methylglyoxal (MGO), a highly reactive dicarbonyl compound, has been implicated in the pathogenesis of type 2 diabetes, vascular complications of diabetes, and several other age-related chronic inflammatory diseases such as cardiovascular disease, cancer, and disorders of the central nervous system. MGO is mainly formed as a byproduct of glycolysis and, under physiological circumstances, detoxified by the glyoxalase system. MGO is the major precursor of nonenzymatic glycation of proteins and DNA, subsequently leading to the formation of advanced glycation end products (AGEs). MGO and MGO-derived AGEs can impact on organs and tissues affecting their functions and structure. In this review we summarize the formation of MGO, the detoxification of MGO by the glyoxalase system, and the biochemical pathways through which MGO is linked to the development of diabetes, vascular complications of diabetes, and other age-related diseases. Although interventions to treat MGO-associated complications are not yet available in the clinical setting, several strategies to lower MGO have been developed over the years. We will summarize several new directions to target MGO stress including glyoxalase inducers and MGO scavengers. Targeting MGO burden may provide new therapeutic applications to mitigate diseases in which MGO plays a crucial role.
Collapse
Affiliation(s)
- C. G. Schalkwijk
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - C. D. A. Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
95
|
Psoralea corylifolia L. Seed Extract Attenuates Methylglyoxal-Induced Insulin Resistance by Inhibition of Advanced Glycation End Product Formation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4310319. [PMID: 31976027 PMCID: PMC6954480 DOI: 10.1155/2019/4310319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/31/2019] [Accepted: 12/05/2019] [Indexed: 01/11/2023]
Abstract
Accumulation of advanced glycation end products (AGEs) in the body has been implicated in the pathogenesis of metabolic conditions, such as diabetes mellitus. Methylglyoxal (MGO), a major precursor of AGEs, has been reported to induce insulin resistance in both in vitro and in vivo studies. Psoralea corylifolia seeds (PCS) have been used as a traditional medicine for several diseases, but their potential application in treating insulin resistance has not yet been evaluated. This study is aimed at investigating whether PCS extract could attenuate insulin resistance induced by MGO. Male C57BL/6N mice (6 weeks old) were administered 1% MGO in their drinking water for 18 weeks, and the PCS extract (200 or 500 mg/kg) was orally administered daily from the first day of the MGO administration. We observed that both 200 and 500 mg/kg PCS extract treatment significantly improved glucose tolerance and insulin sensitivity and markedly restored p-Akt and p-IRS1/2 expression in the livers of the MGO-administered mice. Additionally, the PCS extract significantly increased the phosphorylation of Akt and IRS-1/2 and glucose uptake in MGO-treated HepG2 cells. Further studies showed that the PCS extract inhibited MGO-induced AGE formation in the HepG2 cells and in the sera of MGO-administered mice. PCS extract also increased the expression of glyoxalase 1 (GLO1) in the liver tissue of MGO-administered mice. The PCS extract significantly decreased the phosphorylation of ERK, p38, and NF-κB and suppressed the mRNA expression of proinflammatory molecules including TNF-α and IL-1β and iNOS in MGO-administered mice. Additionally, we demonstrated that the PCS extract attenuated oxidative stress, as evidenced by the reduced ROS production in the MGO-treated cells and the enhanced expression of antioxidant enzymes in the liver of MGO-administered mice. Thus, PCS extract ameliorated the MGO-induced insulin resistance in HepG2 cells and in mice by reducing oxidative stress via the inhibition of AGE formation. These findings suggest the potential of PCS extract as a candidate for the prevention and treatment of insulin resistance.
Collapse
|
96
|
Jiang T, Gu J, Chen W, Chang Q. Resveratrol inhibits high-glucose-induced inflammatory "metabolic memory" in human retinal vascular endothelial cells through SIRT1-dependent signaling. Can J Physiol Pharmacol 2019; 97:1141-1151. [PMID: 31638409 DOI: 10.1139/cjpp-2019-0201] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Diabetes induces vascular endothelial damage and this study investigated high-glucose-induced inflammation "metabolic memory" of human retinal vascular endothelial cells (HRVECs), the effects of resveratrol on HRVECs, and the underlying signaling. HRVECs were grown under various conditions and assayed for levels of sirtuin 1 (SIRT1); acetylated nuclear factor κB (Ac-NF-κB); NOD-like receptor family, pyrin domain containing 3 (NLRP3); and other inflammatory cytokines; and cell viability. A high glucose concentration induced HRVEC inflammation metabolic memory by decreasing SIRT1 and increasing Ac-NF-κB, NLRP3, caspase 1, interleukin-1β, inducible nitric oxide synthase, and tumor necrosis factor α, whereas exposure of HRVECs to a high glucose medium for 4 days, followed by a normal glucose concentration for an additional 4 days, failed to reverse these changes. A high glucose concentration also significantly reduced HRVEC viability. In contrast, resveratrol, a selective SIRT1 activator, markedly enhanced HRVEC viability and reduced the inflammatory cytokines expressions. In addition, high glucose reduced AMP-activated protein kinase (AMPK) phosphorylation and retained during the 4 days of the reversal period of culture. The effects of resveratrol were abrogated after co-treatment with the SIRT1 inhibitor nicotinamide and the AMPK inhibitor compound C. In conclusion, resveratrol was able to reverse high-glucose-induced inflammation "metabolic memory" of HRVECs by activation of the SIRT1/AMPK/NF-κB pathway.
Collapse
Affiliation(s)
- Tingting Jiang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.,Key Laboratory of Myopia of National Health Commission, Fudan University, Shanghai 200000, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai 200000, China
| | - Junxiang Gu
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.,Key Laboratory of Myopia of National Health Commission, Fudan University, Shanghai 200000, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai 200000, China
| | - Wenwen Chen
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.,Key Laboratory of Myopia of National Health Commission, Fudan University, Shanghai 200000, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai 200000, China
| | - Qing Chang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.,Key Laboratory of Myopia of National Health Commission, Fudan University, Shanghai 200000, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai 200000, China
| |
Collapse
|
97
|
Li H, Zheng L, Chen C, Liu X, Zhang W. Brain Senescence Caused by Elevated Levels of Reactive Metabolite Methylglyoxal on D-Galactose-Induced Aging Mice. Front Neurosci 2019; 13:1004. [PMID: 31619960 PMCID: PMC6760031 DOI: 10.3389/fnins.2019.01004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/04/2019] [Indexed: 12/29/2022] Open
Abstract
Aging is a complex natural phenomenon that is manifested by degenerative changes in the structure and function of cells and tissues. D-Galactose-induced aging mice are an artificial accelerated aging model that causes memory and learning impairment, oxidative stress, and neuroinflammation. In this study, we examined the underlying mechanism of an aging mouse model induced by D-galactose. Our behavioral Morris water maze results revealed that D-galactose administration for 2 months significantly induced memory and learning impairment in C57BL/6J mice. High performance liquid chromatography (HPLC) results showed elevated levels of the metabolite methylglyoxal (MG) in D-galactose-induced aging mice. Whether and how D-galactose induces senescence by elevated levels of reactive metabolite MG remain unclear. In our study, MG mainly accumulated through the following two aspects: to increase its source, namely, the triose phosphate produced by the glycolysis pathway, and to reduce its detoxification system, namely, the glyoxalase system. Therefore, elevated MG levels may be one of the causes of brain senescence in D-galactose-induced mice. However, the molecular mechanism of the increased level of the reaction metabolite methylglyoxal requires further exploration.
Collapse
Affiliation(s)
- Hong Li
- Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing, China.,Engineering Research Center of Natural Medicine, Faculty of Geographical Science, Ministry of Education, Beijing Normal University, Beijing, China
| | - Ling Zheng
- Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing, China.,Engineering Research Center of Natural Medicine, Faculty of Geographical Science, Ministry of Education, Beijing Normal University, Beijing, China
| | - Chao Chen
- The Laboratory of Vector Biology, College of Engineering, Beijing Normal University, Zhuhai, China
| | - Xiaoli Liu
- National and Local United Engineering Research Center for Panax Notoginseng Resources Protection and Utilization Technology, Kunming, China
| | - Wensheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing, China.,Engineering Research Center of Natural Medicine, Faculty of Geographical Science, Ministry of Education, Beijing Normal University, Beijing, China.,The Laboratory of Vector Biology, College of Engineering, Beijing Normal University, Zhuhai, China.,National and Local United Engineering Research Center for Panax Notoginseng Resources Protection and Utilization Technology, Kunming, China
| |
Collapse
|
98
|
Wolf AR, Wesener DA, Cheng J, Houston-Ludlam AN, Beller ZW, Hibberd MC, Giannone RJ, Peters SL, Hettich RL, Leyn SA, Rodionov DA, Osterman AL, Gordon JI. Bioremediation of a Common Product of Food Processing by a Human Gut Bacterium. Cell Host Microbe 2019; 26:463-477.e8. [PMID: 31585844 DOI: 10.1016/j.chom.2019.09.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/23/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Dramatic increases in processed food consumption represent a global health threat. Maillard reaction products (MRPs), which are common in processed foods, form upon heat-induced reaction of amino acids with reducing sugars and include advanced glycation end products with deleterious health effects. To examine how processed foods affect the microbiota, we fed gnotobiotic mice, colonized with 54 phylogenetically diverse human gut bacterial strains, defined sugar-rich diets containing whey as the protein source or a matched amino acid mixture. Whey or ϵ-fructoselysine, an MRP in whey and many processed foods, selectively increases Collinsella intestinalis absolute abundance and induces Collinsella expression of genomic loci directing import and metabolism of ϵ-fructoselysine to innocuous products. This locus is repressed by glucose in C. aerofaciens, whose abundance decreases with whey, but is not repressed in C. intestinalis. Identifying gut organisms responding to and degrading potentially harmful processed food components has implications for food science, microbiome science, and public health.
Collapse
Affiliation(s)
- Ashley R Wolf
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Darryl A Wesener
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jiye Cheng
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Alexandra N Houston-Ludlam
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Zachary W Beller
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Matthew C Hibberd
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Richard J Giannone
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Samantha L Peters
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Robert L Hettich
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Semen A Leyn
- A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127994, Russia; Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dmitry A Rodionov
- A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127994, Russia; Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
99
|
Zhuang A, Yap FYT, McCarthy D, Leung C, Sourris KC, Penfold SA, Thallas-Bonke V, Coughlan MT, Schulz BL, Forbes JM. Globally elevating the AGE clearance receptor, OST48, does not protect against the development of diabetic kidney disease, despite improving insulin secretion. Sci Rep 2019; 9:13664. [PMID: 31541173 PMCID: PMC6754370 DOI: 10.1038/s41598-019-50221-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
The accumulation of advanced glycation end products (AGEs) have been implicated in the development and progression of diabetic kidney disease (DKD). There has been interest in investigating the potential of AGE clearance receptors, such as oligosaccharyltransferase-48 kDa subunit (OST48) to prevent the detrimental effects of excess AGE accumulation seen in the diabetic kidney. Here the objective of the study was to increase the expression of OST48 to examine if this slowed the development of DKD by facilitating the clearance of AGEs. Groups of 8-week-old heterozygous knock-in male mice (n = 9-12/group) over-expressing the gene encoding for OST48, dolichyl-diphosphooligosaccharide-protein glycosyltransferase (DDOST+/-) and litter mate controls were randomised to either (i) no diabetes or (ii) diabetes induced via multiple low-dose streptozotocin and followed for 24 weeks. By the study end, global over expression of OST48 increased glomerular OST48. This facilitated greater renal excretion of AGEs but did not affect circulating or renal AGE concentrations. Diabetes resulted in kidney damage including lower glomerular filtration rate, albuminuria, glomerulosclerosis and tubulointerstitial fibrosis. In diabetic mice, tubulointerstitial fibrosis was further exacerbated by global increases in OST48. There was significantly insulin effectiveness, increased acute insulin secretion, fasting insulin concentrations and AUCinsulin observed during glucose tolerance testing in diabetic mice with global elevations in OST48 when compared to diabetic wild-type littermates. Overall, this study suggested that despite facilitating urinary-renal AGE clearance, there were no benefits observed on kidney functional and structural parameters in diabetes afforded by globally increasing OST48 expression. However, the improvements in insulin secretion seen in diabetic mice with global over-expression of OST48 and their dissociation from effects on kidney function warrant future investigation.
Collapse
Affiliation(s)
- Aowen Zhuang
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia.,School of Medicine, University of Queensland, St Lucia, 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
| | - Domenica McCarthy
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Chris Leung
- Department of Medicine, University of Melbourne, Austin Hospital, Heidelberg, Australia
| | - Karly C Sourris
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Sally A Penfold
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | | | | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Josephine M Forbes
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia. .,School of Medicine, University of Queensland, St Lucia, Australia. .,Mater Clinical School, University of Queensland, St Lucia, Australia.
| |
Collapse
|
100
|
Park SJ, Nam J, Ahn CW, Kim Y. Anti-diabetic properties of different fractions of Korean red ginseng. JOURNAL OF ETHNOPHARMACOLOGY 2019; 236:220-230. [PMID: 30849506 DOI: 10.1016/j.jep.2019.01.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Korean red ginseng (KRG) has been traditionally used to treat diabetes. Ginsenosides are considered as the major bioactive components mediating anti-diabetic effects of KRG. However, considering that ginsenosides account for only about 3-4% of ginsengs, other fractions of KRG may also carry potential anti-diabetic effects. There is no study reporting the differentiated effects of ginsenosides (Spn) and non-saponin fractions (NSpn) of KRG on glycemic control. AIM OF THE STUDY We investigated the effects of KRG, Spn, and NSpn on the indications of glycemic control and sought to elucidate physiological factors contributing their effects. MATERIALS AND METHODS Human T2DM mimicking Nagoya-Shibata-Yasuda (NSY/hos) mice were given KRG, Spn, or NSpn admixed in rodent diet at 200 mg/kg/day for 24 weeks. Glycemic and obesity indications, blood lipid profile, systematic and local oxidative stress markers in metabolically important organs, and systematic inflammatory markers were assessed. Molecular assessments associated with glycemic control in liver and skeletal muscle were further performed. RESULTS KRG attenuated deterioration in glucose homeostasis as evidenced by significantly lower fasting blood glucose from 22nd week and AUC during GTT at the end of the experiment compare to control. Spn enhanced insulin secretion in response to glucose stimulation and reduced protein level of glycogen phosphorylase in liver. On the other hand, NSpn ameliorated oxidative stress and inflammation. Some beneficial effects of Spn and NSpn were reflected in KRG treated mice. KRG also attenuated the accumulation of malondialdehyde in skeletal muscle and, accordingly, enhanced insulin responsiveness compare to control. CONCLUSION Anti-diabetic properties of KRG are not solely determined by the contents of ginsenosides but the harmonic functions of its different fractions.
Collapse
Affiliation(s)
- Soo Jeong Park
- Brain Korea 21 Plus Project for Medical Science, Yonsei University, 50-1 Yonsei-Ro Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| | - Jisun Nam
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, 50-1 Yonsei-Ro Seodaemun-Gu, Seoul, 03722, Republic of Korea; Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-Ro Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| | - Chul Woo Ahn
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, 50-1 Yonsei-Ro Seodaemun-Gu, Seoul, 03722, Republic of Korea; Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-Ro Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| | - YuSik Kim
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, 50-1 Yonsei-Ro Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|