1
|
Sroga GE, Vashishth D. In vivo glycation-interplay between oxidant and carbonyl stress in bone. JBMR Plus 2024; 8:ziae110. [PMID: 39386996 PMCID: PMC11458925 DOI: 10.1093/jbmrpl/ziae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 06/18/2024] [Accepted: 07/28/2024] [Indexed: 10/12/2024] Open
Abstract
Metabolic syndromes (eg, obesity, type 2 diabetes (T2D), atherosclerosis, and neurodegenerative diseases) and aging, they all have a strong component of carbonyl and reductive-oxidative (redox) stress. Reactive carbonyl (RCS) and oxidant (ROS) stress species are commonly generated as products or byproducts of cellular metabolism or are derived from the environment. RCS and ROS can play a dual role in living organisms. Some RCS and ROS function as signaling molecules, which control cellular defenses against biological and environmental assaults. However, due to their high reactivity, RCS and ROS inadvertently interact with different cellular and extracellular components, which can lead to the formation of undesired posttranslational modifications of bone matrix proteins. These are advanced glycation (AGEs) and glycoxidation (AGOEs) end products generated in vivo by non-enzymatic amino-carbonyl reactions. In this review, metabolic processes involved in generation of AGEs and AGOEs within and on protein surfaces including extracellular bone matrix are discussed from the perspective of cellular metabolism and biochemistry of certain metabolic syndromes. The impact of AGEs and AGOEs on some characteristics of mineral is also discussed. Different therapeutic approaches with the potential to prevent the formation of RCS, ROS, and the resulting formation of AGEs and AGOEs driven by these chemicals are also briefly reviewed. These are antioxidants, scavenging agents of reactive species, and newly emerging technologies for the development of synthetic detoxifying systems. Further research in the area of in vivo glycation and glycoxidation should lead to the development of diverse new strategies for halting the progression of metabolic complications before irreversible damage to body tissues materializes.
Collapse
Affiliation(s)
- Grażyna E Sroga
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Shirley Ann Jackson PhD Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Deepak Vashishth
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Shirley Ann Jackson PhD Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Engineering and Precision Medicine, Rensselaer-Icahn School of Medicine at Mount Sinai, 619 West 54th Street, New York, NY 10019, United States
| |
Collapse
|
2
|
Uceda AB, Mariño L, Casasnovas R, Adrover M. An overview on glycation: molecular mechanisms, impact on proteins, pathogenesis, and inhibition. Biophys Rev 2024; 16:189-218. [PMID: 38737201 PMCID: PMC11078917 DOI: 10.1007/s12551-024-01188-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 05/14/2024] Open
Abstract
The formation of a heterogeneous set of advanced glycation end products (AGEs) is the final outcome of a non-enzymatic process that occurs in vivo on long-life biomolecules. This process, known as glycation, starts with the reaction between reducing sugars, or their autoxidation products, with the amino groups of proteins, DNA, or lipids, thus gaining relevance under hyperglycemic conditions. Once AGEs are formed, they might affect the biological function of the biomacromolecule and, therefore, induce the development of pathophysiological events. In fact, the accumulation of AGEs has been pointed as a triggering factor of obesity, diabetes-related diseases, coronary artery disease, neurological disorders, or chronic renal failure, among others. Given the deleterious consequences of glycation, evolution has designed endogenous mechanisms to undo glycation or to prevent it. In addition, many exogenous molecules have also emerged as powerful glycation inhibitors. This review aims to provide an overview on what glycation is. It starts by explaining the similarities and differences between glycation and glycosylation. Then, it describes in detail the molecular mechanism underlying glycation reactions, and the bio-molecular targets with higher propensity to be glycated. Next, it discusses the precise effects of glycation on protein structure, function, and aggregation, and how computational chemistry has provided insights on these aspects. Finally, it reports the most prevalent diseases induced by glycation, and the endogenous mechanisms and the current therapeutic interventions against it.
Collapse
Affiliation(s)
- Ana Belén Uceda
- Departament de Química, Universitat de Les Illes Balears, Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa Km 7.5, 07122 Palma, Spain
| | - Laura Mariño
- Departament de Química, Universitat de Les Illes Balears, Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa Km 7.5, 07122 Palma, Spain
| | - Rodrigo Casasnovas
- Departament de Química, Universitat de Les Illes Balears, Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa Km 7.5, 07122 Palma, Spain
| | - Miquel Adrover
- Departament de Química, Universitat de Les Illes Balears, Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa Km 7.5, 07122 Palma, Spain
| |
Collapse
|
3
|
Ivanov SV, Rose KL, Colon S, Hudson BG, Bhave G, Voziyan P. Mechanism of peroxidasin inactivation in hyperglycemia: Heme damage by reactive oxygen species. Biochem Biophys Res Commun 2023; 689:149237. [PMID: 37984175 PMCID: PMC10702573 DOI: 10.1016/j.bbrc.2023.149237] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023]
Abstract
Diabetic complications present a serious health problem. Functional damage to proteins due to post-translational modifications by glycoxidation reactions is a known factor contributing to pathology. Extracellular proteins are especially vulnerable to diabetic damage because robust antioxidant defenses are lacking outside the cell. We investigated glucose-induced inactivation of peroxidasin (PXDN), a heme protein catalyzing sulfilimine crosslinking of collagen IV that reinforce the basement membranes (BM). Experiments using physiological diabetic glucose levels were carried out to exclude several potential mechanisms of PXDN inactivation i.e., direct adduction of glucose, reactive carbonyl damage, steric hindrance, and osmotic stress. Further experiments established that PXDN activity was inhibited via heme degradation by reactive oxygen species. Activity of another extracellular heme protein, myeloperoxidase, was unaffected by glucose because its heme was resistant to glucose-induced oxidative degradation. Our findings point to specific mechanisms which may compromise BM structure and stability in diabetes and suggest potential modes of protection.
Collapse
Affiliation(s)
- Sergey V Ivanov
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Selene Colon
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Billy G Hudson
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Gautam Bhave
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37212, USA; Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Paul Voziyan
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA; Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
4
|
Mossine VV, Mawhinney TP. 1-Amino-1-deoxy-d-fructose ("fructosamine") and its derivatives. Adv Carbohydr Chem Biochem 2023; 83:27-132. [PMID: 37968038 DOI: 10.1016/bs.accb.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Fructosamine has long been considered as a key intermediate of the Maillard reaction, which to a large extent is responsible for specific aroma, taste, and color formation in thermally processed or dehydrated foods. Since the 1980s, however, as a product of the Amadori rearrangement reaction between glucose and biologically significant amines such as proteins, fructosamine has experienced a boom in biomedical research, mainly due to its relevance to pathologies in diabetes and aging. In this chapter, we assess the scope of the knowledge on and applications of fructosamine-related molecules in chemistry, food, and health sciences, as reflected mostly in publications within the past decade. Methods of fructosamine synthesis and analysis, its chemical, and biological properties, and degradation reactions, together with fructosamine-modifying and -recognizing proteins are surveyed.
Collapse
Affiliation(s)
- Valeri V Mossine
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Thomas P Mawhinney
- Department of Biochemistry, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
5
|
Menta PR, Neves RC, Machado VS. Association of time to metritis diagnosis with circulating concentration of metabolites, minerals, and haptoglobin in Jersey cows. J Dairy Sci 2023; 106:5029-5042. [PMID: 37268564 DOI: 10.3168/jds.2022-22979] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/30/2023] [Indexed: 06/04/2023]
Abstract
Our objective was to investigate the association of early metritis [EMET, diagnosed at <5 d in milk (DIM)] and late metritis (LMET, diagnosed at ≥5 DIM) with circulating concentrations of energy metabolites, minerals, and haptoglobin (Hp) throughout the first 14 d postpartum. A total of 379 purebred Jersey cows were enrolled in a prospective cohort study from a single herd in west Texas. Cows were examined for metritis using the Metricheck device (Simcro Ltd.) at 4, 7, and 10 DIM. Cows identified by farm employees as possible metritis cases were also evaluated for metritis. Blood samples were collected for analysis of concentrations of Ca, Mg, and glucose at DIM 1 through 5, 7, 10, and 14. Albumin, urea, fructosamine, free fatty acids (FFA), creatinine, and β-hydroxybutyrate (BHB) were analyzed at DIM 3, 5, 7, 10, and 14, and Hp at DIM 1 through 5 and 7. Data were analyzed using the MIXED and PHREG procedures of SAS (SAS Institute Inc.). A series of mixed general linear models accounting for repeated measures were fitted to the data. The independent variables metritis [no metritis (NMET), EMET, and LMET], DIM of analyte assessment, and parity were forced in all models. Multivariable Cox proportional hazard models were built to assess the risk of pregnancy and culling within 150 DIM. The overall metritis incidence was 26.9% (EMET = 49; LMET = 53; NMET = 277). Average concentrations of glucose, Mg, and urea were not associated with metritis. The associations of Ca, creatinine, BHB, and fructosamine with metritis were dependent on the DIM of analyte assessment. Cows categorized as EMET and LMET had, on average, lower albumin and fructosamine compared with NMET cows. Both EMET and LMET cows had, on average, greater BHB than NMET cows. A greater FFA concentration was only observed in cows diagnosed with EMET compared with NMET cows (EMET = 0.58, LMET = 0.52, NMET = 0.48 mmol/L). Additionally, circulating Hp concentration was greater for LMET and EMET compared with NMET cows, and EMET cows had greater Hp compared with LMET cows (EMET = 1.15; LMET = 1.00; NMET = 0.84). In conclusion, several blood biomarkers were temporally associated with early versus late metritis diagnosis in postpartum Jersey cows. No meaningful differences were observed in production, reproduction, or culling between EMET and LMET cows. These results suggest that cows with EMET undergo a more severe degree of inflammation and negative energy balance compared with NMET cows.
Collapse
Affiliation(s)
- P R Menta
- Department of Veterinary Sciences, Texas Tech University, Lubbock 79415
| | - R C Neves
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN 47907
| | - V S Machado
- Department of Veterinary Sciences, Texas Tech University, Lubbock 79415.
| |
Collapse
|
6
|
Song H, Ma H, Shi J, Liu Y, Kan C, Hou N, Han J, Sun X, Qiu H. Optimizing glycation control in diabetes: An integrated approach for inhibiting nonenzymatic glycation reactions of biological macromolecules. Int J Biol Macromol 2023:125148. [PMID: 37268079 DOI: 10.1016/j.ijbiomac.2023.125148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/16/2023] [Accepted: 05/27/2023] [Indexed: 06/04/2023]
Abstract
Diabetes is a multifactorial disorder that increases mortality and disability due to its complications. A key driver of these complications is nonenzymatic glycation, which generates advanced glycation end-products (AGEs) that impair tissue function. Therefore, effective nonenzymatic glycation prevention and control strategies are urgently needed. This review comprehensively describes the molecular mechanisms and pathological consequences of nonenzymatic glycation in diabetes and outlines various anti-glycation strategies, such as lowering plasma glucose, interfering with the glycation reaction, and degrading early and late glycation products. Diet, exercise, and hypoglycemic medications can reduce the onset of high glucose at the source. Glucose or amino acid analogs such as flavonoids, lysine and aminoguanidine competitively bind to proteins or glucose to block the initial nonenzymatic glycation reaction. In addition, deglycation enzymes such as amadoriase, fructosamine-3-kinase, parkinson's disease protein, glutamine amidotransferase-like class 1 domain-containing 3A and terminal FraB deglycase can eliminate existing nonenzymatic glycation products. These strategies involve nutritional, pharmacological, and enzymatic interventions that target different stages of nonenzymatic glycation. This review also emphasizes the therapeutic potential of anti-glycation drugs for preventing and treating diabetes complications.
Collapse
Affiliation(s)
- Hongwei Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongyan Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yongping Liu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jing Han
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261053, Shandong, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| |
Collapse
|
7
|
Kim Y. Blood and Tissue Advanced Glycation End Products as Determinants of Cardiometabolic Disorders Focusing on Human Studies. Nutrients 2023; 15:nu15082002. [PMID: 37111220 PMCID: PMC10144557 DOI: 10.3390/nu15082002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiometabolic disorders are characterised by a cluster of interactive risk determinants such as increases in blood glucose, lipids and body weight, as well as elevated inflammation and oxidative stress and gut microbiome changes. These disorders are associated with onset of type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). T2DM is strongly associated with CVD. Dietary advanced glycation end products (dAGEs) attributable from modern diets high in sugar and/or fat, highly processed foods and high heat-treated foods can contribute to metabolic etiologies of cardiometabolic disorders. This mini review aims to determine whether blood dAGEs levels and tissue dAGEs levels are determinants of the prevalence of cardiometabolic disorders through recent human studies. ELISA (enzyme-linked immunosorbent assay), high-performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS) for blood dAGEs measurement and skin auto fluorescence (SAF) for skin AGEs measurement can be used. Recent human studies support that a diet high in AGEs can negatively influence glucose control, body weight, blood lipid levels and vascular health through the elevated oxidative stress, inflammation, blood pressure and endothelial dysfunction compared with a diet low in AGEs. Limited human studies suggested a diet high in AGEs could negatively alter gut microbiota. SAF could be considered as one of the predictors affecting risks for cardiometabolic disorders. More intervention studies are needed to determine how dAGEs are associated with the prevalence of cardiometabolic disorders through gut microbiota changes. Further human studies are conducted to find the association between CVD events, CVD mortality and total mortality through SAF measurement, and a consensus on whether tissue dAGEs act as a predictor of CVD is required.
Collapse
Affiliation(s)
- Yoona Kim
- Department of Food and Nutrition, Institute of Agriculture and Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
8
|
Advanced Glycation End-Products and Diabetic Neuropathy of the Retina. Int J Mol Sci 2023; 24:ijms24032927. [PMID: 36769249 PMCID: PMC9917392 DOI: 10.3390/ijms24032927] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Diabetic retinopathy is a tissue-specific neurovascular impairment of the retina in patients with both type 1 and type 2 diabetes. Several pathological factors are involved in the progressive impairment of the interdependence between cells that consist of the neurovascular units (NVUs). The advanced glycation end-products (AGEs) are one of the major pathological factors that cause the impairments of neurovascular coupling in diabetic retinopathy. Although the exact mechanisms for the toxicities of the AGEs in diabetic retinopathy have not been definitively determined, the AGE-receptor of the AGE (RAGE) axis, production of reactive oxygen species, inflammatory reactions, and the activation of the cell death pathways are associated with the impairment of the NVUs in diabetic retinopathy. More specifically, neuronal cell death is an irreversible change that is directly associated with vision reduction in diabetic patients. Thus, neuroprotective therapies must be established for diabetic retinopathy. The AGEs are one of the therapeutic targets to examine to ameliorate the pathological changes in the NVUs in diabetic retinopathy. This review focuses on the basic and pathological findings of AGE-induced neurovascular abnormalities and the potential therapeutic approaches, including the use of anti-glycated drugs to protect the AGE-induced impairments of the NVUs in diabetic retinopathy.
Collapse
|
9
|
Huo S, Wang Q, Shi W, Peng L, Jiang Y, Zhu M, Guo J, Peng D, Wang M, Men L, Huang B, Lv J, Lin L. ATF3/SPI1/SLC31A1 Signaling Promotes Cuproptosis Induced by Advanced Glycosylation End Products in Diabetic Myocardial Injury. Int J Mol Sci 2023; 24:ijms24021667. [PMID: 36675183 PMCID: PMC9862315 DOI: 10.3390/ijms24021667] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Cuproptosis resulting from copper (Cu) overload has not yet been investigated in diabetic cardiomyopathy (DCM). Advanced glycosylation end products (AGEs) induced by persistent hyperglycemia play an essential role in cardiotoxicity. To clarify whether cuproptosis was involved in AGEs-induced cardiotoxicity, we analyzed the toxicity of AGEs and copper in AC16 cardiomyocytes and in STZ-induced or db/db-diabetic mouse models. The results showed that copper ionophore elesclomol induced cuproptosis in cardiomyocytes. It was only rescued by copper chelator tetrathiomolybdate rather than by other cell death inhibitors. Intriguingly, AGEs triggered cardiomyocyte death and aggravated it when incubated with CuCl2 or elesclomol-CuCl2. Moreover, AGEs increased intracellular copper accumulation and exhibited features of cuproptosis, including loss of Fe-S cluster proteins (FDX1, LIAS, NDUFS8 and ACO2) and decreased lipoylation of DLAT and DLST. These effects were accompanied by decreased mitochondrial oxidative respiration, including downregulated mitochondrial respiratory chain complex, decreased ATP production and suppressed mitochondrial complex I and III activity. Additionally, AGEs promoted the upregulation of copper importer SLC31A1. We predicted that ATF3 and/or SPI1 might be transcriptional factors of SLC31A1 by online databases and validated that by ATF3/SPI1 overexpression. In diabetic mice, copper and AGEs increases in the blood and heart were observed and accompanied by cardiac dysfunction. The protein and mRNA profile changes in diabetic hearts were consistent with cuproptosis. Our findings showed, for the first time, that excessive AGEs and copper in diabetes upregulated ATF3/SPI1/SLC31A1 signaling, thereby disturbing copper homeostasis and promoting cuproptosis. Collectively, the novel mechanism might be an alternative potential therapeutic target for DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jiagao Lv
- Correspondence: (J.L.); or (L.L.); Tel.: +86-13971600239 (J.L.); +86-18971097627 (L.L.)
| | - Li Lin
- Correspondence: (J.L.); or (L.L.); Tel.: +86-13971600239 (J.L.); +86-18971097627 (L.L.)
| |
Collapse
|
10
|
Role of Nitric Oxide-Derived Metabolites in Reactions of Methylglyoxal with Lysine and Lysine-Rich Protein Leghemoglobin. Int J Mol Sci 2022; 24:ijms24010168. [PMID: 36613614 PMCID: PMC9820652 DOI: 10.3390/ijms24010168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Carbonyl stress occurs when reactive carbonyl compounds (RCC), such as reducing sugars, dicarbonyls etc., accumulate in the organism. The interaction of RCC carbonyl groups with amino groups of molecules is called the Maillard reaction. One of the most active RCCs is α-dicarbonyl methylglyoxal (MG) that modifies biomolecules forming non-enzymatic glycation products. Organic free radicals are formed in the reaction between MG and lysine or Nα-acetyllysine. S-nitrosothiols and nitric oxide (•NO) donor PAPA NONOate increased the yield of organic free radical intermediates, while other •NO-derived metabolites, namely, nitroxyl anion and dinitrosyl iron complexes (DNICs) decreased it. At the late stages of the Maillard reaction, S-nitrosoglutathione (GSNO) also inhibited the formation of glycation end products (AGEs). The formation of a new type of DNICs, bound with Maillard reaction products, was found. The results obtained were used to explain the glycation features of legume hemoglobin-leghemoglobin (Lb), which is a lysine-rich protein. In Lb, lysine residues can form fluorescent cross-linked AGEs, and •NO-derived metabolites slow down their formation. The knowledge of these processes can be used to increase the stability of Lb. It can help in better understanding the impact of stress factors on legume plants and contribute to the production of recombinant Lb for biotechnology.
Collapse
|
11
|
Willett TL, Voziyan P, Nyman JS. Causative or associative: A critical review of the role of advanced glycation end-products in bone fragility. Bone 2022; 163:116485. [PMID: 35798196 PMCID: PMC10062699 DOI: 10.1016/j.bone.2022.116485] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/02/2022]
Abstract
The accumulation of advanced glycation end-products (AGEs) in the organic matrix of bone with aging and chronic disease such as diabetes is thought to increase fracture risk independently of bone mass. However, to date, there has not been a clinical trial to determine whether inhibiting the accumulation of AGEs is effective in preventing low-energy, fragility fractures. Moreover, unlike with cardiovascular or kidney disease, there are also no pre-clinical studies demonstrating that AGE inhibitors or breakers can prevent the age- or diabetes-related decrease in the ability of bone to resist fracture. In this review, we critically examine the case for a long-standing hypothesis that AGE accumulation in bone tissue degrades the toughening mechanisms by which bone resists fracture. Prior research into the role of AGEs in bone has primarily measured pentosidine, an AGE crosslink, or bulk fluorescence of hydrolysates of bone. While significant correlations exist between these measurements and mechanical properties of bone, multiple AGEs are both non-fluorescent and non-crosslinking. Since clinical studies are equivocal on whether circulating pentosidine is an indicator of elevated fracture risk, there needs to be a more complete understanding of the different types of AGEs including non-crosslinking adducts and multiple non-enzymatic crosslinks in bone extracellular matrix and their specific contributions to hindering fracture resistance (biophysical and biological). By doing so, effective strategies to target AGE accumulation in bone with minimal side effects could be investigated in pre-clinical and clinical studies that aim to prevent fragility fractures in conditions that bone mass is not the underlying culprit.
Collapse
Affiliation(s)
- Thomas L Willett
- Biomedical Engineering Program, Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada.
| | - Paul Voziyan
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffry S Nyman
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
12
|
Walnut Prevents Cognitive Impairment by Regulating the Synaptic and Mitochondrial Dysfunction via JNK Signaling and Apoptosis Pathway in High-Fat Diet-Induced C57BL/6 Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165316. [PMID: 36014555 PMCID: PMC9414791 DOI: 10.3390/molecules27165316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/30/2022]
Abstract
This study was conducted to evaluate the protective effect of Juglans regia (walnut, Gimcheon 1ho cultivar, GC) on high-fat diet (HFD)-induced cognitive dysfunction in C57BL/6 mice. The main physiological compounds of GC were identified as pedunculagin/casuariin isomer, strictinin, tellimagrandin I, ellagic acid-O-pentoside, and ellagic acid were identified using UPLC Q-TOF/MS analysis. To evaluate the neuro-protective effect of GC, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 2′,7′-dichlorodihydrofluorecein diacetate (DCF-DA) analysis were conducted in H2O2 and high glucose-induced neuronal PC12 cells and hippocampal HT22 cells. GC presented significant cell viability and inhibition of reactive oxygen species (ROS) production. GC ameliorated behavioral and memory dysfunction through Y-maze, passive avoidance, and Morris water maze tests. In addition, GC reduced white adipose tissue (WAT), liver fat mass, and serum dyslipidemia. To assess the inhibitory effect of antioxidant system deficit, lipid peroxidation, ferric reducing antioxidant power (FRAP), and advanced glycation end products (AGEs) were conducted. Administration of GC protected the antioxidant damage against HFD-induced diabetic oxidative stress. To estimate the ameliorating effect of GC, acetylcholine (ACh) level, acetylcholinesterase (AChE) activity, and expression of AChE and choline acetyltransferase (ChAT) were conducted, and the supplements of GC suppressed the cholinergic system impairment. Furthermore, GC restored mitochondrial dysfunction by regulating the mitochondrial ROS production and mitochondrial membrane potential (MMP) levels in cerebral tissues. Finally, GC ameliorated cerebral damage by synergically regulating the protein expression of the JNK signaling and apoptosis pathway. These findings suggest that GC could provide a potential functional food source to improve diabetic cognitive deficits and neuronal impairments.
Collapse
|
13
|
Avenues for post-translational protein modification prevention and therapy. Mol Aspects Med 2022; 86:101083. [PMID: 35227517 PMCID: PMC9378364 DOI: 10.1016/j.mam.2022.101083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 12/22/2022]
Abstract
Non-enzymatic post-translational modifications (nPTMs) of proteins have emerged as novel risk factors for the genesis and progression of various diseases. We now have a variety of experimental and established therapeutic strategies to target harmful nPTMs and potentially improve clinical outcomes. Protein carbamylation and glycation are two common and representative nPTMs that have gained considerable attention lately as favorable therapeutic targets with emerging clinical evidence. Protein carbamylation is associated with the occurrence of cardiovascular disease (CVD) and mortality in patients with chronic kidney disease (CKD); and advanced glycation end products (AGEs), a heterogeneous group of molecules produced in a series of glycation reactions, have been linked to various diabetic complications. Therefore, reducing the burden of protein carbamylation and AGEs is an appealing and promising therapeutic approach. This review chapter summarizes potential anti-nPTM therapy options in CKD, CVD, and diabetes along with clinical implications. Using two prime examples-protein carbamylation and AGEs-we discuss the varied preventative and therapeutic options to mitigate these pathologic nPTMs in detail. We provide in-depth case studies on carbamylation in the setting of kidney disease and AGEs in metabolic disorders, with an emphasis on the relevance to reducing adverse clinical outcomes such as CKD progression, cardiovascular events, and mortality. Overall, whether specific efforts to lower carbamylation and AGE burden will yield definitive clinical improvement in humans remains largely to be seen. However, the scientific rationale for such pursuits is demonstrated herein.
Collapse
|
14
|
Bazdar P, Jalalvand AR, Akbari V, Khodarahmi R, Goicoechea HC. Resolving interactions of miglitol with normal and glycated human serum albumin by multivariate methods. Anal Biochem 2021; 630:114339. [PMID: 34411552 DOI: 10.1016/j.ab.2021.114339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 02/05/2023]
Abstract
This article reports results of one of our projects related to the investigation of interactions of miglitol (MIG) with normal human serum albumin (HSA) and glycated HSA (GHSA) with the help of recording spectroscopic and electrochemical data. The experimental data were analyzed by conventional and chemometric methods to extract useful information for comprehensive justifications of the interactions of the MIG with HSA and GHSA. Hard- and soft-modeling chemometric methods were used to extract quantitative and qualitative information. Then, molecular docking techniques were used to further investigation of the binding of the MIG with HSA and GHSA and the extracted results were compatible with those obtained by experimental methods. Finally, according to the binding of the BV with HSA and GHSA, second-order differential pulse voltammetric data were recorded and calibrated with three-way calibration methods for exploiting second-order advantage for determination of the GHSA in the presence of the HSA to develop a novel chemometrics assisted-electroanalytical method for diagnostic and monitoring of diabetic.
Collapse
Affiliation(s)
- Parsa Bazdar
- Research Institute Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali R Jalalvand
- Research Center of Oils and Fats, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Vali Akbari
- Research Center of Oils and Fats, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hector C Goicoechea
- Laboratorio de Desarrollo Analítico y Quimiometría (LADAQ), Cátedra de Química Analítica I, Universidad Nacional del Litoral, Ciudad Universitaria, CC 242, S3000ZAA, Santa Fe, Argentina
| |
Collapse
|
15
|
Velichkova S, Foubert K, Pieters L. Natural Products as a Source of Inspiration for Novel Inhibitors of Advanced Glycation Endproducts (AGEs) Formation. PLANTA MEDICA 2021; 87:780-801. [PMID: 34341977 DOI: 10.1055/a-1527-7611] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Protein glycation, a post-translational modification found in biological systems, is often associated with a core defect in glucose metabolism. In particular, advanced glycation endproducts are complex heterogeneous sugar-derived protein modifications implicated in the progression of pathological conditions such as atherosclerosis, diabetic complications, skin diseases, rheumatism, hypertension, and neurodegenerative diseases. Undoubtedly, there is the need to expand the knowledge about antiglycation agents that can offer a therapeutic approach in preventing and treating health issues of high social and economic importance. Although various compounds have been under consideration, little data from clinical trials are available, and there is a lack of approved and registered antiglycation agents. Next to the search for novel synthetic advanced glycation endproduct inhibitors, more and more the efforts of scientists are focusing on researching antiglycation compounds from natural origin. The main purpose of this review is to provide a thorough overview of the state of scientific knowledge in the field of natural products from plant origin (e.g., extracts and pure compounds) as inhibitors of advanced glycation endproduct formation in the period between 1990 and 2019. Moreover, the objectives of the summary also include basic chemistry of AGEs formation and classification, pathophysiological significance of AGEs, mechanisms for inhibiting AGEs formation, and examples of several synthetic anti-AGEs drugs.
Collapse
Affiliation(s)
- Stefaniya Velichkova
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Kenn Foubert
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Luc Pieters
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
16
|
Qiu HY, Hou NN, Shi JF, Liu YP, Kan CX, Han F, Sun XD. Comprehensive overview of human serum albumin glycation in diabetes mellitus. World J Diabetes 2021; 12:1057-1069. [PMID: 34326954 PMCID: PMC8311477 DOI: 10.4239/wjd.v12.i7.1057] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/06/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
The presence of excess glucose in blood is regarded as a sweet hurt for patients with diabetes. Human serum albumin (HSA) is the most abundant protein in human plasma, which undergoes severe non-enzymatic glycation with glucose in patients with diabetes; this modifies the structure and function of HSA. Furthermore, the advanced glycation end products produced by glycated HSA can cause pathological damage to the human body through various signaling pathways, eventually leading to complications of diabetes. Many potential glycation sites on HSA have different degrees of sensitivity to glucose concentration. This review provides a comprehensive assessment of the in vivo glycation sites of HSA; it also discusses the effects of glycation on the structure and function of HSA. Moreover, it addresses the relationship between HSA glycation and diabetes complications. Finally, it focuses on the value of non-enzymatic glycation of HSA in diabetes-related clinical applications.
Collapse
Affiliation(s)
- Hong-Yan Qiu
- Department of Endocrinology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Ning-Ning Hou
- Department of Endocrinology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Jun-Feng Shi
- Department of Endocrinology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Yong-Ping Liu
- Department of Endocrinology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Cheng-Xia Kan
- Department of Endocrinology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Fang Han
- Department of Pathology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Xiao-Dong Sun
- Department of Endocrinology, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| |
Collapse
|
17
|
Wiglusz K, Żurawska-Płaksej E, Rorbach-Dolata A, Piwowar A. How Does Glycation Affect Binding Parameters of the Albumin-Gliclazide System in the Presence of Drugs Commonly Used in Diabetes? In Vitro Spectroscopic Study. Molecules 2021; 26:molecules26133869. [PMID: 34202801 PMCID: PMC8270297 DOI: 10.3390/molecules26133869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/16/2022] Open
Abstract
In this research, the selected drugs commonly used in diabetes and its comorbidities (gliclazide, cilazapril, atorvastatin, and acetylsalicylic acid) were studied for their interactions with bovine serum albumin-native and glycated. Two different spectroscopic methods, fluorescence quenching and circular dichroism, were utilized to elucidate the binding interactions of the investigational drugs. The glycation process was induced in BSA by glucose and was confirmed by the presence of advanced glycosylation end products (AGEs). The interaction between albumin and gliclazide, with the presence of another drug, was confirmed by calculation of association constants (0.11-1.07 × 104 M-1). The nature of changes in the secondary structure of a protein depends on the drug used and the degree of glycation. Therefore, these interactions may have an influence on pharmacokinetic parameters.
Collapse
Affiliation(s)
- Katarzyna Wiglusz
- Department of Analytical Chemistry, Wroclaw Medical University, Borowska 211, PL-50556 Wrocław, Poland;
| | - Ewa Żurawska-Płaksej
- Department of Toxicology, Wroclaw Medical University, Borowska 211, PL-50556 Wrocław, Poland; (A.R.-D.); (A.P.)
- Department of Pharmaceutical Biochemistry, Wroclaw Medical University, Borowska 211, PL-50556 Wrocław, Poland
- Correspondence: ; Tel.: +48-71-784-0453
| | - Anna Rorbach-Dolata
- Department of Toxicology, Wroclaw Medical University, Borowska 211, PL-50556 Wrocław, Poland; (A.R.-D.); (A.P.)
| | - Agnieszka Piwowar
- Department of Toxicology, Wroclaw Medical University, Borowska 211, PL-50556 Wrocław, Poland; (A.R.-D.); (A.P.)
| |
Collapse
|
18
|
Sant S, Wang D, Abidi M, Walker G, Ferrell N. Mechanical characterization of native and sugar-modified decellularized kidneys. J Mech Behav Biomed Mater 2021; 114:104220. [PMID: 33257205 PMCID: PMC7855467 DOI: 10.1016/j.jmbbm.2020.104220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/28/2020] [Accepted: 11/20/2020] [Indexed: 10/22/2022]
Abstract
Decellularized organs have the potential to be used as scaffolds for tissue engineering organ replacements. The mechanical properties of the extracellular matrix (ECM) following decellularization are critical for structural integrity and for regulation of cell function upon recellularization. Advanced glycation end products (AGEs) accumulate in the ECM with age and their formation is accelerated by several pathological conditions including diabetes. Some AGEs span multiple amino acids to form crosslinks that may alter the mechanical properties of the ECM. The goal of this work was to evaluate how sugar-induced modifications to the ECM affect the mechanical behavior of decellularized kidney. The compressive and tensile properties of the kidney ECM were evaluated using an accelerated model of AGE formation by ribose. Results show that ribose modifications significantly alter the mechanical behavior of decellularized kidney. Increased resistance to deformation corresponds to increased ECM crosslinking, and mechanical changes can be partially mitigated by AGE inhibition. The degree of post-translational modification of the ECM is dependent on the age and health of the organ donor and may play a role in regulating the mechanical properties of decellularized organs.
Collapse
Affiliation(s)
- Snehal Sant
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Dan Wang
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Minhal Abidi
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Gwyneth Walker
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Nicholas Ferrell
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States; Department of Biomedical Engineering, Vanderbilt University, United States; Vanderbilt Center for Kidney Disease, United States.
| |
Collapse
|
19
|
Bagherzadeh-Yazdi M, Bohlooli M, Khajeh M, Ghamari F, Ghaffari-Moghaddam M, Poormolaie N, Khatibi A, Hasanein P, Sheibani N. Acetoacetate enhancement of glucose mediated DNA glycation. Biochem Biophys Rep 2020; 25:100878. [PMID: 33364448 PMCID: PMC7750490 DOI: 10.1016/j.bbrep.2020.100878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 11/05/2020] [Accepted: 12/09/2020] [Indexed: 11/17/2022] Open
Abstract
Acetoacetate (AA) is a ketone body, which generates reactive oxygen species (ROS). ROS production is impacted by the formation of covalent bonds between amino groups of biomacromolecules and reducing sugars (glycation). Glycation can damage DNA by causing strand breaks, mutations, and changes in gene expression. DNA damage could contribute to the pathogenesis of various diseases, including neurological disorders, complications of diabetes, and aging. Here we studied the enhancement of glucose-mediated DNA glycation by AA for the first time. The effect of AA on the structural changes, Amadori and advanced glycation end products (AGEs) formation of DNA incubated with glucose for 4 weeks were investigated using various techniques. These included UV-Vis, circular dichroism (CD) and fluorescence spectroscopy, and agarose gel electrophoresis. The results of UV-Vis and fluorescence spectroscopy confirmed that AA increased the DNA-AGE formation. The NBT test showed that AA also increased Amadori product formation of glycated DNA. Based on the CD and agarose gel electrophoresis results, the structural changes of glycated DNA was increased in the presence of AA. The chemiluminescence results indicated that AA increased ROS formation. Thus AA has an activator role in DNA glycation, which could enhance the adverse effects of glycation under high glucose conditions.
Collapse
Affiliation(s)
| | - M Bohlooli
- Department of Biology, University of Zabol, Zabol, Iran
| | - M Khajeh
- Department of Chemistry, University of Zabol, Zabol, Iran
| | - F Ghamari
- Department of Biology, Payame Noor University, Ghazvin, Iran
| | | | - N Poormolaie
- Department of Chemistry, University of Zabol, Zabol, Iran
| | - A Khatibi
- Department of Biotechnology, Alzahra University, Tehran, Iran
| | - P Hasanein
- Department of Biology, University of Zabol, Zabol, Iran
| | - N Sheibani
- Departments of Ophthalmology and Visual Sciences, Cell and Regenerative Biology, and Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
20
|
Li J, Jeong SY, Xiong B, Tseng A, Mahon AB, Isaacman S, Gordeuk VR, Cho J. Repurposing pyridoxamine for therapeutic intervention of intravascular cell-cell interactions in mouse models of sickle cell disease. Haematologica 2020; 105:2407-2419. [PMID: 33054081 PMCID: PMC7556679 DOI: 10.3324/haematol.2019.226720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/29/2019] [Indexed: 12/25/2022] Open
Abstract
Adherent neutrophils on vascular endothelium positively contribute to cell-cell aggregation and vaso-occlusion in sickle cell disease. In the present study, we demonstrated that pyridoxamine, a derivative of vitamin B6, might be a therapeutic agent to alleviate intravascular cell-cell aggregation in sickle cell disease. Using real-time intravital microscopy, we found that one oral administration of pyridoxamine dose-dependently increased the rolling influx of neutrophils and reduced neutrophil adhesion to endothelial cells in cremaster microvessels of sickle cell disease mice challenged with hypoxia-reoxygenation. Short-term treatment also mitigated neutrophil-endothelial cell and neutrophil-platelet interactions in the microvessels and improved the survival of sickle cell disease mice challenged with tumor necrosis factor-α. The inhibitory effects of pyridoxamine on intravascular cell-cell interactions were potentiated by co-treatment with hydroxyurea. We observed that long-term (5.5 months) oral treatment with pyridoxamine significantly diminished the adhesive function of neutrophils and platelets and down-regulated the expression of E-selectin and intercellular adhesion molecule-1 on the vascular endothelium in tumor necrosis factor-α-challenged sickle cell disease mice. Ex vivo studies revealed that the surface amount of αMβ2 integrin was significantly decreased in stimulated neutrophils isolated from sickle cell disease mice treated with pyridoxamine-containing water. Studies using platelets and neutrophils from sickle cell disease mice and patients suggested that treatment with pyridoxamine reduced the activation state of platelets and neutrophils. These results suggest that pyridoxamine may be a novel therapeutic and a supplement to hydroxyurea to prevent and treat vaco-occlusion events in sickle cell disease.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Si-Yeon Jeong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Bei Xiong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Alan Tseng
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | | | - Victor R. Gordeuk
- Section of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Comprehensive Sickle Cell Center, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| |
Collapse
|
21
|
Sant S, Wang D, Agarwal R, Dillender S, Ferrell N. Glycation alters the mechanical behavior of kidney extracellular matrix. Matrix Biol Plus 2020; 8:100035. [PMID: 33543034 PMCID: PMC7852306 DOI: 10.1016/j.mbplus.2020.100035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022] Open
Abstract
The mechanical properties of the extracellular matrix (ECM) are important in maintaining normal physiological function, and changes in ECM mechanics drive disease. The biochemical structure of the ECM is modified with aging and in diseases such as diabetes. One mechanism of ECM modification is the non-enzymatic reaction between sugars and ECM proteins resulting in formation of advanced glycation end products (AGEs). Some AGE reactions result in formation of molecular crosslinks within or between matrix proteins, but it is not clear how sugar-mediated biochemical modification of the ECM translates to changes in kidney ECM mechanical properties. AGE-mediated changes in ECM mechanics may have pathological consequences in diabetic kidney disease. To determine how sugars alter the mechanical properties of the kidney ECM, we employ custom methodologies to evaluate the mechanical properties of isolated tubular basement membrane (TBM) and glomerular ECM. Results show that the mechanical properties of TBM and glomerular ECM stiffness were altered by incubation in glucose and ribose. Mechanical behavior of TBM and glomerular ECM were further evaluated using mechanical models for hyperelastic materials in tension and compression. Increased ECM stiffness following sugar modification corresponded to increased crosslinking as determined by ECM fluorescence and reduced pepsin extractability of sugar modified ECM. These results show that sugar-induced modifications significantly affect the mechanical properties of kidney ECM. AGE-mediated changes in ECM mechanics may be important in progression of chronic diseases including diabetic kidney disease.
Collapse
Affiliation(s)
- Snehal Sant
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States of America
| | - Dan Wang
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States of America
| | - Rishabh Agarwal
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States of America
| | - Sarah Dillender
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States of America
| | - Nicholas Ferrell
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States of America.,Department of Biomedical Engineering, Vanderbilt University, United States of America.,Vanderbilt Center for Kidney Disease, United States of America
| |
Collapse
|
22
|
Study of glycation process of human carbonic anhydrase II as well as investigation concerning inhibitory influence of 3-beta-hydroxybutyrate on it. Int J Biol Macromol 2020; 149:443-449. [PMID: 31978481 DOI: 10.1016/j.ijbiomac.2020.01.192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/12/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Glycation is a non-enzymatic reaction between carbonyl groups in sugar and free amino groups in proteins. This reaction leads to changes in structure and functions of proteins in which the advanced glycation end products (AGEs) are the final outcome and cause many complications in diabetic patients. We herein examined the effect of fasting on the glycation process of human Carbonic anhydrase II under physiological conditions (37 °C and pH 7.4) employing various techniques, including Ultraviolet-visible spectroscopy, fluorescence spectroscopy and CD Spectroscopy. We found an increased 3-beta-hydroxybutyrate upon fasting. We studied various samples of control carbonic anhydrase (without glucose and 3-beta-hydroxybutyrate), carbonic anhydrase with glucose, carbonic anhydrase treated with 3-beta-hydroxybutyrate (BHB) and carbonic anhydrase along with glucose and 3-beta-hydroxybutyrate. The samples were incubated for 35 days under physiological conditions. Our results indicated that 3-beta-hydroxybutyrate inhibited the glycation process, decreased glucose binding to the protein, prevented the formation of AGEs, and modified the enzyme activity. Our findings would open new windows toward the enzymatic procedure which would have profound implication in understanding the diabetes mechanisms.
Collapse
|
23
|
Sarmah S, Pahari S, Das S, Belwal VK, Jana M, Singha Roy A. Non-enzymatic glycation of human serum albumin modulates its binding efficacy towards bioactive flavonoid chrysin: A detailed study using multi-spectroscopic and computational methods. J Biomol Struct Dyn 2020; 39:476-492. [PMID: 31900044 DOI: 10.1080/07391102.2019.1711196] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The non-enzymatic glycation of plasma proteins by reducing sugars have important consequences on the conformational and functional properties of protein. The formation of advanced glycation end products (AGEs) is responsible for cell death and other pathological conditions. We have synthesized the glycated human serum albumin (gHSA) and characterized the same by using differential spectroscopic measurements. The aim of the present study is to determine the effect of glycation on the binding of human serum albumin (HSA) with bioactive flavonoid chrysin, which possesses anti-cancer, anti-inflammatory and anti-oxidant activities. The interaction of chrysin with HSA and gHSA was studied using multi-spectroscopic, molecular docking and molecular dynamics (MD) simulation techniques. Chrysin quenched the intrinsic fluorescence of both HSA and gHSA by static quenching mechanism. The value of the binding constant (Kb) for the interaction of HSA-chrysin complex (4.779 ± 0.623 × 105 M-1 at 300 K) was found to be higher than that of gHSA-chrysin complex (2.206 ± 0.234 × 105 M-1 at 300 K). Hence, non-enzymatic glycation of HSA significantly reduced its binding affinity towards chrysin. The % α-helicity of HSA was found to get enhanced upon binding with chrysin, and minimal changes were observed for the gHSA-chrysin complex. Site marker probe studies indicated that chrysin binds to subdomain IIA and IIIA of both HSA and gHSA. The results from molecular docking and MD simulation studies correlated well with the experimental findings. Electrostatic interactions followed by hydrogen bonding and hydrophobic interactions played major roles in the binding process. These observations may have some useful insights into the field of pharmaceutics.
Collapse
Affiliation(s)
- Sharat Sarmah
- Department of Chemistry, National Institute of Technology, Meghalaya, Shillong, India
| | - Somdev Pahari
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela, India
| | - Sourav Das
- Department of Chemistry, National Institute of Technology, Meghalaya, Shillong, India
| | - Vinay Kumar Belwal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Madhurima Jana
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela, India
| | - Atanu Singha Roy
- Department of Chemistry, National Institute of Technology, Meghalaya, Shillong, India
| |
Collapse
|
24
|
Ramis R, Ortega-Castro J, Caballero C, Casasnovas R, Cerrillo A, Vilanova B, Adrover M, Frau J. How Does Pyridoxamine Inhibit the Formation of Advanced Glycation End Products? The Role of Its Primary Antioxidant Activity. Antioxidants (Basel) 2019; 8:E344. [PMID: 31480509 PMCID: PMC6770850 DOI: 10.3390/antiox8090344] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 12/28/2022] Open
Abstract
Pyridoxamine, one of the natural forms of vitamin B6, is known to be an effective inhibitor of the formation of advanced glycation end products (AGEs), which are closely related to various human diseases. Pyridoxamine forms stable complexes with metal ions that catalyze the oxidative reactions taking place in the advanced stages of the protein glycation cascade. It also reacts with reactive carbonyl compounds generated as byproducts of protein glycation, thereby preventing further protein damage. We applied Density Functional Theory to study the primary antioxidant activity of pyridoxamine towards three oxygen-centered radicals (•OOH, •OOCH3 and •OCH3) to find out whether this activity may also play a crucial role in the context of protein glycation inhibition. Our results show that, at physiological pH, pyridoxamine can trap the •OCH3 radical, in both aqueous and lipidic media, with rate constants in the diffusion limit (>1.0 × 108 M - 1 s - 1 ). The quickest pathways involve the transfer of the hydrogen atoms from the protonated pyridine nitrogen, the protonated amino group or the phenolic group. Its reactivity towards •OOH and •OOCH3 is smaller, but pyridoxamine can still scavenge them with moderate rate constants in aqueous media. Since reactive oxygen species are also involved in the formation of AGEs, these results highlight that the antioxidant capacity of pyridoxamine is also relevant to explain its inhibitory role on the glycation process.
Collapse
Affiliation(s)
- Rafael Ramis
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Joaquín Ortega-Castro
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain.
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain.
| | - Carmen Caballero
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Rodrigo Casasnovas
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Antonia Cerrillo
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Bartolomé Vilanova
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Miquel Adrover
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Juan Frau
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| |
Collapse
|
25
|
Tagliazucchi D, Martini S, Conte A. Protocatechuic and 3,4-Dihydroxyphenylacetic Acids Inhibit Protein Glycation by Binding Lysine through a Metal-Catalyzed Oxidative Mechanism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:7821-7831. [PMID: 31260293 DOI: 10.1021/acs.jafc.9b02357] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The mechanism of inhibition of advanced glycation end product (AGE) formation by protocatechuic acid and 3,4-dihydroxyphenylacetic acid (DHPA) has been studied using a widespread applied in vitro model system composed of bovine serum albumin (BSA) and supraphysiological glucose concentrations. Protocatechuic acid and DHPA inhibited the formation of Amadori compounds, fluorescent AGEs (IC50 = 62.1 ± 1.4 and 155.4 ± 1.1 μmol/L, respectively), and Nε-(carboxymethyl)lysine (IC50 = 535.3 ± 1.1 and 751.2 ± 1.0 μmol/L, respectively). BSA was pretreated with the two phenolic acids, and the formation of BSA-phenolic acid adducts was estimated by nanoflow liquid chromatography-electrospray ionization-quadrupole time-of-flight mass spectrometry. Results showed that the tested phenolic acids bound key sites of glycation in BSA through a metal-catalyzed oxidative mechanism. The antiglycative activity mechanism involved the formation of BSA-phenolic acid adducts, and it is unlikely that this occurs in vivo. These results raise the problem to design in vitro models closer to physiological conditions to reach biologically sound conclusions.
Collapse
Affiliation(s)
- Davide Tagliazucchi
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Amendola 2 , 42100 Reggio Emilia , Italy
| | - Serena Martini
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Amendola 2 , 42100 Reggio Emilia , Italy
| | - Angela Conte
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Amendola 2 , 42100 Reggio Emilia , Italy
| |
Collapse
|
26
|
Jud P, Sourij H. Therapeutic options to reduce advanced glycation end products in patients with diabetes mellitus: A review. Diabetes Res Clin Pract 2019; 148:54-63. [PMID: 30500546 DOI: 10.1016/j.diabres.2018.11.016] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/14/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus (DM) defines metabolic disorders, characterised by elevated levels of blood glucose. Chronic hyperglycaemic state promotes consequently the formation of advanced glycation end products (AGEs) and the expression of their receptor (RAGE) which aggravate many diabetic complications. Due to the relevant role of AGEs and RAGE, several therapeutic approaches with an anti-AGE or RAGE-antagonizing effect are investigated. These therapeutic options include AGE cross-link breakers, AGE inhibitors, RAGE antagonists, drugs clinically approved for various indications like antidiabetic, antihypertensive drugs or statins, as well as dietary and phytotherapeutic approaches. The aim of this review is to give an overview of these therapeutic approaches, their outcomes in clinical studies and their role in the management of diabetes and its complications.
Collapse
Affiliation(s)
- Philipp Jud
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
27
|
Abdullah K, Qais FA, Ahmad I, Hasan H, Naseem I. Study of pyridoxamine against glycation and reactive oxygen species production in human serum albumin as model protein: An in vitro & ex vivo approach. Int J Biol Macromol 2018; 120:1734-1743. [DOI: 10.1016/j.ijbiomac.2018.09.176] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/26/2018] [Accepted: 09/26/2018] [Indexed: 12/23/2022]
|
28
|
Bohlooli M, Ghaffari-Moghaddam M, Khajeh M, Sheibani N. Determination of Amadori Product in Glycated Human Serum Albumin by Spectroscopy Methods. ChemistrySelect 2018. [DOI: 10.1002/slct.201800207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | | | - Mostafa Khajeh
- Department of Chemistry; University of Zabol; Zabol Iran
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences and Biomedical Engineering, University of Wisconsin; School of Medicine and Public Health; Madison, WI USA
| |
Collapse
|
29
|
Sanchis P, Rivera R, Berga F, Fortuny R, Adrover M, Costa-Bauza A, Grases F, Masmiquel L. Phytate Decreases Formation of Advanced Glycation End-Products in Patients with Type II Diabetes: Randomized Crossover Trial. Sci Rep 2018; 8:9619. [PMID: 29941991 PMCID: PMC6018557 DOI: 10.1038/s41598-018-27853-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/12/2018] [Indexed: 01/13/2023] Open
Abstract
Myo-inositol hexaphosphate (phytate; IP6) is a natural compound that is abundant in cereals, legumes, and nuts and it has the ability to chelate metal cations. The binding of IP6 to transition metals suggests that it could be used for the treatment of metal-catalyzed protein glycation, which appears to trigger diabetes-related diseases. Our in vitro studies showed that IP6 reduced the formation of Fe3+-catalyzed advanced glycation end-products (AGEs). This led us to perform a randomized cross-over trial to investigate the impact of the daily consumption IP6 on protein glycation in patients with type 2 diabetes mellitus (T2DM; n = 33). Thus, we measured AGEs, glycated hemoglobin (HbA1c), several vascular risk factors, and urinary IP6 at baseline and at the end of the intervention period. Patients who consumed IP6 supplements for 3 months had lower levels of circulating AGEs and HbA1c than those who did not consume IP6. This is the first report to show that consumption of IP6 inhibits protein glycation in patients with T2DM. Considering that AGEs contribute to microvascular and macrovascular complications in T2DM, our data indicates that dietary supplementation with IP6 should be considered as a therapy to prevent the formation of AGEs and therefore, the development of diabetes-related diseases in patients with T2DM.
Collapse
Affiliation(s)
- Pilar Sanchis
- Endocrinology Department, Research Unit, Hospital Son Llàtzer, Institute of Health Sciences Research [IUNICS- IdISBa], 07198, Palma of Mallorca, Spain.
- Laboratory of Renal Lithiasis Research, University of Balearic Islands, Institute of Health Sciences Research [IUNICS- IdISBa], 07122, Palma of Mallorca, Spain.
| | - Rosmeri Rivera
- Endocrinology Department, Research Unit, Hospital Son Llàtzer, Institute of Health Sciences Research [IUNICS- IdISBa], 07198, Palma of Mallorca, Spain
| | - Francisco Berga
- Laboratory of Renal Lithiasis Research, University of Balearic Islands, Institute of Health Sciences Research [IUNICS- IdISBa], 07122, Palma of Mallorca, Spain
| | - Regina Fortuny
- Laboratory Department, Hospital Son Llàtzer, 07198, Palma of Mallorca, Spain
| | - Miquel Adrover
- Department of Chemistry, University of Balearic Islands, Ctra. Valldemossa km 7.5, 07122, Palma of Mallorca, Spain
| | - Antonia Costa-Bauza
- Laboratory of Renal Lithiasis Research, University of Balearic Islands, Institute of Health Sciences Research [IUNICS- IdISBa], 07122, Palma of Mallorca, Spain
| | - Felix Grases
- Laboratory of Renal Lithiasis Research, University of Balearic Islands, Institute of Health Sciences Research [IUNICS- IdISBa], 07122, Palma of Mallorca, Spain
| | - Luis Masmiquel
- Endocrinology Department, Research Unit, Hospital Son Llàtzer, Institute of Health Sciences Research [IUNICS- IdISBa], 07198, Palma of Mallorca, Spain.
| |
Collapse
|
30
|
Non-enzymatic glycation enhances human serum albumin binding capacity to sodium fluorescein at room temperature: A spectroscopic analysis. Clin Chim Acta 2017; 469:180-186. [DOI: 10.1016/j.cca.2017.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 02/08/2023]
|
31
|
Dicarbonyls and Advanced Glycation End-Products in the Development of Diabetic Complications and Targets for Intervention. Int J Mol Sci 2017; 18:ijms18050984. [PMID: 28475116 PMCID: PMC5454897 DOI: 10.3390/ijms18050984] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/11/2017] [Accepted: 05/02/2017] [Indexed: 01/17/2023] Open
Abstract
Advanced glycation end-products (AGEs) are non-enzymatic protein and amino acid adducts as well as DNA adducts which form from dicarbonyls and glucose. AGE formation is enhanced in diabetes and is associated with the development of diabetic complications. In the current review, we discuss mechanisms that lead to enhanced AGE levels in the context of diabetes and diabetic complications. The methylglyoxal-detoxifying glyoxalase system as well as alternative pathways of AGE detoxification are summarized. Therapeutic approaches to interfere with different pathways of AGE formation are presented.
Collapse
|
32
|
Kumar Pasupulati A, Chitra PS, Reddy GB. Advanced glycation end products mediated cellular and molecular events in the pathology of diabetic nephropathy. Biomol Concepts 2016; 7:293-309. [DOI: 10.1515/bmc-2016-0021] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/18/2016] [Indexed: 01/11/2023] Open
Abstract
AbstractDiabetic nephropathy (DN) is a major cause of morbidity and mortality in diabetic patients and a leading cause of end-stage renal disease (ESRD). Degenerative changes such as glomerular hypertrophy, hyperfiltration, widening of basement membranes, tubulointerstitial fibrosis, glomerulosclerosis and podocytopathy manifest in various degrees of proteinuria in DN. One of the key mechanisms implicated in the pathogenesis of DN is non-enzymatic glycation (NEG). NEG is the irreversible attachment of reducing sugars onto free amino groups of proteins by a series of events, which include the formation of Schiff’s base and an Amadori product to yield advanced glycation end products (AGEs). AGE modification of client proteins from the extracellular matrix induces crosslinking, which is often associated with thickening of the basement membrane. AGEs activate several intracellular signaling cascades upon interaction with receptor for AGEs (RAGE), which manifest in aberrant cellular responses such as inflammation, apoptosis and autophagy, whereas other receptors such as AGE-R1, AGE-R3 and scavenger receptors also bind to AGEs and ensue endocytosis and degradation of AGEs. Elevated levels of both serum and tissue AGEs are associated with adverse renal outcome. Increased evidence supports that attenuation of AGE formation and/or inhibition of RAGE activation manifest(s) in improving renal function. This review provides insights of NEG, discusses the cellular and molecular events triggered by AGEs, which manifest in the pathogenesis of DN including renal fibrosis, podocyte epithelial-mesenchymal transition and activation of renin-angiotensin system. Therapies designed to target AGEs, such as inhibitors of AGEs formation and crosslink breakers, are discussed.
Collapse
Affiliation(s)
| | - P. Swathi Chitra
- 2Department of Biochemistry, National Institute of Nutrition, Tarnaka, Hyderabad 500 007, India
| | - G. Bhanuprakash Reddy
- 2Department of Biochemistry, National Institute of Nutrition, Tarnaka, Hyderabad 500 007, India
| |
Collapse
|
33
|
Shumaev KB, Kosmachevskaya OV, Nasybullina EI, Gromov SV, Novikov AA, Topunov AF. New dinitrosyl iron complexes bound with physiologically active dipeptide carnosine. J Biol Inorg Chem 2016; 22:153-160. [PMID: 27878396 DOI: 10.1007/s00775-016-1418-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/15/2016] [Indexed: 01/08/2023]
Abstract
Dinitrosyl iron complexes (DNICs) are physiological NO derivatives and account for many NO functions in biology. Polyfunctional dipeptide carnosine (beta-alanyl-L-histidine) is considered to be a very promising pharmacological agent. It was shown that in the system containing carnosine, iron ions and Angeli's salt, a new type of DNICs bound with carnosine as ligand {(carnosine)2-Fe-(NO)2}, was formed. We studied how the carbonyl compound methylglyoxal influenced this process. Carnosine-bound DNICs appear to be one of the cell's adaptation mechanisms when the amount of reactive carbonyl compounds increases at hyperglycemia. These complexes can also participate in signal and regulatory ways of NO and can act as protectors at oxidative and carbonyl stress conditions.
Collapse
Affiliation(s)
- Konstantin B Shumaev
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Prospect 33, Moscow, 119071, Russian Federation
| | - Olga V Kosmachevskaya
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Prospect 33, Moscow, 119071, Russian Federation
| | - Elvira I Nasybullina
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Prospect 33, Moscow, 119071, Russian Federation
| | - Sergey V Gromov
- National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Alexander A Novikov
- National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Alexey F Topunov
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Prospect 33, Moscow, 119071, Russian Federation.
| |
Collapse
|
34
|
The role of acetoacetate in Amadori product formation of human serum albumin. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 163:345-51. [PMID: 27614245 DOI: 10.1016/j.jphotobiol.2016.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/02/2016] [Accepted: 09/03/2016] [Indexed: 11/20/2022]
Abstract
Amadori product is an important and stable intermediate, which is produced during glycation process. It is a marker of hyperglycemia in diabetes mellitus, and its accumulation in the body contributes to microvascular complication of diabetes including diabetic nephropathy and retinopathy. In this study, the effect of acetoacetate on the formation of Amadori products and biophysical properties of human serum albumin (HSA), after incubation with glucose, was investigated using various methods. These included circular dichroism (CD), Fourier transform infrared (FTIR) spectroscopy, and UV-visible and fluorescence spectroscopy. Our results indicated that the production of Amadori products in HSA incubated with glucose (GHSA) was increased in the presence of acetoacetate. We also detected alterations in the secondary and tertiary structure of GHSA, which was increased in the presence of acetoacetate. These changes were attributed to the formation of covalent bonds between the carbonyl group of acetoacetate and the nucleophilic groups (lysine residues) of HSA. Thus, acetoacetate can enhance the production of Amadori products through formation of covalent bonds with biomaterials.
Collapse
|
35
|
Targeting advanced glycation with pharmaceutical agents: where are we now? Glycoconj J 2016; 33:653-70. [PMID: 27392438 DOI: 10.1007/s10719-016-9691-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/11/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023]
Abstract
Advanced glycation end products (AGEs) are the final products of the Maillard reaction, a complex process that has been studied by food chemists for a century. Over the past 30 years, the biological significance of advanced glycation has also been discovered. There is mounting evidence that advanced glycation plays a homeostatic role within the body and that food-related Maillard products, intermediates such as reactive α-dicarbonyl compounds and AGEs, may influence this process. It remains to be understood, at what point AGEs and their intermediates become pathogenic and contribute to the pathogenesis of chronic diseases that inflict current society. Diabetes and its complications have been a major focus of AGE biology due to the abundance of excess sugar and α-dicarbonyls in this family of diseases. While further temporal information is required, a number of pharmacological agents that inhibit components of the advanced glycation pathway have already showed promising results in preclinical models. These therapies appear to have a wide range of mechanistic actions to reduce AGE load. Some of these agents including Alagebrium, have translated successfully to clinical trials, while others such as aminoguanidine, have had undesirable side-effect profiles. This review will discuss different pharmacological agents that have been used to reduce AGE burden in preclinical models of disease with a focus on diabetes and its complications, compare outcomes of those therapies that have reached clinical trials, and provide further rationale for the use of inhibitors of the glycation pathway in chronic diseases.
Collapse
|
36
|
Skrypnyk NI, Voziyan P, Yang H, de Caestecker CR, Theberge MC, Drouin M, Hudson B, Harris RC, de Caestecker MP. Pyridoxamine reduces postinjury fibrosis and improves functional recovery after acute kidney injury. Am J Physiol Renal Physiol 2016; 311:F268-77. [PMID: 27194713 DOI: 10.1152/ajprenal.00056.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) is a common and independent risk factor for death and chronic kidney disease (CKD). Despite promising preclinical data, there is no evidence that antioxidants reduce the severity of injury, increase recovery, or prevent CKD in patients with AKI. Pyridoxamine (PM) is a structural analog of vitamin B6 that interferes with oxidative macromolecular damage via a number of different mechanisms and is in a phase 3 clinical efficacy trial to delay CKD progression in patients with diabetic kidney disease. Because oxidative stress is implicated as one of the main drivers of renal injury after AKI, the ability of PM to interfere with multiple aspects of oxidative damage may be favorable for AKI treatment. In these studies we therefore evaluated PM treatment in a mouse model of AKI. Pretreatment with PM caused a dose-dependent reduction in acute tubular injury, long-term postinjury fibrosis, as well as improved functional recovery after ischemia-reperfusion AKI (IR-AKI). This was associated with a dose-dependent reduction in the oxidative stress marker isofuran-to-F2-isoprostane ratio, indicating that PM reduces renal oxidative damage post-AKI. PM also reduced postinjury fibrosis when administered 24 h after the initiating injury, but this was not associated with improvement in functional recovery after IR-AKI. This is the first report showing that treatment with PM reduces short- and long-term injury, fibrosis, and renal functional recovery after IR-AKI. These preclinical findings suggest that PM, which has a favorable clinical safety profile, holds therapeutic promise for AKI and, most importantly, for prevention of adverse long-term outcomes after AKI.
Collapse
Affiliation(s)
- Nataliya I Skrypnyk
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul Voziyan
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Haichun Yang
- Division of Pathology, Microbiology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christian R de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Billy Hudson
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Biochemistry, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark P de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Cell and Developmental Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; and
| |
Collapse
|
37
|
Bohlooli M, Ghaffari-Moghaddam M, Khajeh M, Aghashiri Z, Sheibani N, Moosavi-Movahedi AA. Acetoacetate promotes the formation of fluorescent advanced glycation end products (AGEs). J Biomol Struct Dyn 2016; 34:2658-2666. [PMID: 26621475 DOI: 10.1080/07391102.2015.1125790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Acetoacetate (AA) is an important ketone body, which produces reactive oxygen species (ROS). Advanced glycation end products (AGEs) are defined as final products of glycation process whose production is influenced by the levels of ROS. The accumulation of AGEs in the body contributes to pathogenesis of many diseases including complications of diabetes, and Alzheimer's and Parkinson's disease. Here, we evaluated the impact of AA on production of AGEs upon incubation of human serum albumin (HSA) with glucose. The effect of AA on the AGEs formation of HSA was studied under physiological conditions after incubation with glucose for 35 days. The physical techniques including circular dichroism (CD) and fluorescence spectroscopy were used to assess the impact of AA on formation and structural changes of glycated HSA (GHSA). Our results indicated that the secondary and tertiary structural changes of GHSA were increased in the presence of AA. The fluorescence intensity measurements of AGEs also showed an increase in AGEs formation. Acetoacetate has an activator effect in formation of AGEs through ROS production. The presence of AA may result in enhanced glycation in the presence of glucose and severity of complications associated with accumulation of AGEs.
Collapse
Affiliation(s)
- Mousa Bohlooli
- a Department of Biology , University of Zabol , Zabol , Iran
| | | | - Mostafa Khajeh
- b Department of Chemistry , University of Zabol , Zabol , Iran
| | - Zohre Aghashiri
- b Department of Chemistry , University of Zabol , Zabol , Iran
| | - Nader Sheibani
- c Departments of Ophthalmology and Visual Sciences and McPherson Eye Research Institute , University of Wisconsin School of Medicine and Public Health , Madison , WI , USA
| | | |
Collapse
|
38
|
Zamani Z, Ghanei M, Panahi Y, Arjmand M, Sadeghi S, Mirkhani F, Parvin S, Salehi M, Sahebkar A, Vahabi F. Serum Metabolomic Profiling of Sulphur Mustard-Exposed Individuals Using (1)H Nuclear Magnetic Resonance Spectroscopy. Basic Clin Pharmacol Toxicol 2015; 118:77-82. [PMID: 26177671 DOI: 10.1111/bcpt.12441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/30/2015] [Indexed: 12/01/2022]
Abstract
Sulphur mustard is an alkylating agent that reacts with different cellular components, causing acute and delayed complications that may remain for decades after exposure. This study aimed to identify differentially expressed metabolites between mustard-exposed individuals suffering from chronic complications compared with unexposed individuals as the control group. Serum samples were obtained from 15 mustard-exposed individuals and 15 apparently healthy unexposed individuals. Metabolomic profiling was performed using (1)H nuclear magnetic resonance spectroscopy, and analyses were carried out using Chenomex and MATLAB softwares. Metabolites were identified using Human Metabolome Database, and the main metabolic pathways were identified using MetaboAnalyst software. Chemometric analysis of serum samples identified 11 differentially expressed metabolites between mustard-exposed and unexposed groups. The main pathways that were influenced by sulphur mustard exposure were related to vitamin B6 (down-regulation), bile acid (up-regulation) and tryptophan (down-regulation) metabolism. Metabolism of vitamin B6, bile acids and tryptophan are the most severely impaired pathways in individuals suffering from chronic mustard-induced complications. These findings may find implications in the monitoring of exposed patients and identification of new therapeutic approaches.
Collapse
Affiliation(s)
- Zahra Zamani
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunus Panahi
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Arjmand
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Sadeghi
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Mirkhani
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| | - Shahram Parvin
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Salehi
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farideh Vahabi
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
39
|
Cao W, Chen J, Chen Y, Chen S, Chen X, Huang H, Liu P. Advanced glycation end products induced immune maturation of dendritic cells controls heart failure through NF-κB signaling pathway. Arch Biochem Biophys 2015; 580:112-20. [PMID: 26163748 DOI: 10.1016/j.abb.2015.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS It is commonly believed that diabetes is an important contributor to heart failure (HF) development. However, the detail effect of diabetogenesis on HF is controversy: both beneficial and harmful roles were reported. In the present study, we aim to explore the unambiguous action of diabetes on chronic HF progression and the underlying mechanism. METHODS Diabetes and myocardial infarction (MI) were induced by streptozotocin (STZ) injection and left-sided thoracotomy and left anterior descending coronary artery (LAD) ligation, respectively. Pyridoxamine was used as the antagonist of advanced glycation end products (AGEs). Adult male SD rats were assigned to 5 groups: Sham; MI; Diabetes (D); Diabetes+MI (DMI) and DMI+pyridoxamine (DMI+P). Animals were sacrificed at the end of 12weeks. The comparison of LV myocardium was made between border zone from MI or DMI animals and control LV tissues from sham-operated animals. Cardiomyocytes and dendritic cells were prepared from the Sprague-Dawley rats and cocultured in the presence or absence of AGEs. RESULTS DMI group showed highest level of AGEs and inflammatory markers, which were significantly reduced in the presence of pyridoxamine. In vitro experiment disclosed AGEs could stimulate DCs differentiation and promote cytokines production, finally upregulated hypertrophy-related genes expression in cardiocytes. Intervention DCs differentiation was sufficient to improve cardiocytes morphology. CONCLUSION Our results clearly demonstrate that diabetes would promote chronic HF progression at least in part through stimulating DCs differentiation and series downstream inflammatory responses induced by AGEs.
Collapse
Affiliation(s)
- Weiwei Cao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Jianwen Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yanfang Chen
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Shaorui Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xi Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Heqing Huang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Engineering Laboratory of Guangdong Province for Druggability and New Drug Evaluation, China
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Engineering Laboratory of Guangdong Province for Druggability and New Drug Evaluation, China.
| |
Collapse
|
40
|
Toth-Manikowski S, Atta MG. Diabetic Kidney Disease: Pathophysiology and Therapeutic Targets. J Diabetes Res 2015; 2015:697010. [PMID: 26064987 PMCID: PMC4430644 DOI: 10.1155/2015/697010] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/17/2015] [Indexed: 12/13/2022] Open
Abstract
Diabetes is a worldwide epidemic that has led to a rise in diabetic kidney disease (DKD). Over the past two decades, there has been significant clarification of the various pathways implicated in the pathogenesis of DKD. Nonetheless, very little has changed in the way clinicians manage patients with this disorder. Indeed, treatment is primarily centered on controlling hyperglycemia and hypertension and inhibiting the renin-angiotensin system. The purpose of this review is to describe the current understanding of how the hemodynamic, metabolic, inflammatory, and alternative pathways are all entangled in pathogenesis of DKD and detail the various therapeutic targets that may one day play a role in quelling this epidemic.
Collapse
Affiliation(s)
- Stephanie Toth-Manikowski
- Division of Nephrology, Johns Hopkins University, 1830 E. Monument Street, Suite 416, Baltimore, MD 21287, USA
| | - Mohamed G. Atta
- Division of Nephrology, Johns Hopkins University, 1830 E. Monument Street, Suite 416, Baltimore, MD 21287, USA
| |
Collapse
|
41
|
Li M, Hagerman AE. Effect of (-)-epigallocatechin-3-gallate on glucose-induced human serum albumin glycation. Free Radic Res 2015; 49:946-53. [PMID: 25794449 DOI: 10.3109/10715762.2015.1016429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
(-)-Epigallocatechin-3-gallate (EGCg) is a naturally occurring polyphenol found in plant-based foods and beverages such as green tea. Although EGCg can eliminate carbonyl species produced by glucose autoxidation and thus can inhibit protein glycation, it is also reported to be a pro-oxidant that stimulates protein glycation in vitro. To better understand the balance between antioxidant and pro-oxidant features of EGCg, we evaluated EGCg-mediated bioactivities in a human serum albumin (HSA)/glucose model by varying three different parameters (glucose level, EGCg concentration, and time of exposure to EGCg). Measurements of glycation-induced fluorescence, protein carbonyls, and electrophoretic mobility showed that the level of HSA glycation was positively related to the glucose level over the range 10-100 mM during a 21-day incubation at 37°C and pH: 7.4. Under mild glycemic pressure (10 mM), long exposure to EGCg enhanced HSA glycation, while brief exposure to low concentrations of EGCg did not. Under high glycemic pressure (100 mM glucose), long exposure to EGCg inhibited glycation. For the first time we showed that brief exposure to EGCg reversed glycation-induced fluorescence, indicating a restorative effect. In conclusion, our research identified glucose level, EGCg concentration, and time of exposure as critical factors dictating EGCg bioactivities in HSA glycation. EGCg did not affect HSA glycation under normal physiological conditions but had a potential therapeutic effect on HSA severely damaged by glycation.
Collapse
Affiliation(s)
- M Li
- Department of Chemistry and Biochemistry, Miami University , Oxford, OH , USA
| | | |
Collapse
|
42
|
Sroga GE, Siddula A, Vashishth D. Glycation of human cortical and cancellous bone captures differences in the formation of Maillard reaction products between glucose and ribose. PLoS One 2015; 10:e0117240. [PMID: 25679213 PMCID: PMC4334514 DOI: 10.1371/journal.pone.0117240] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 12/22/2014] [Indexed: 01/22/2023] Open
Abstract
To better understand some aspects of bone matrix glycation, we used an in vitro glycation approach. Within two weeks, our glycation procedures led to the formation of advanced glycation end products (AGEs) at the levels that corresponded to approx. 25-30 years of the natural in vivo glycation. Cortical and cancellous bones from human tibias were glycated in vitro using either glucose (glucosylation) or ribose (ribosylation). Both glucosylation and ribosylation led to the formation of higher levels of AGEs and pentosidine (PEN) in cancellous than cortical bone dissected from all tested donors (young, middle-age and elderly men and women). More efficient glycation of bone matrix proteins in cancellous bone most likely depended on the higher porosity of this tissue, which facilitated better accessibility of the sugars to the matrix proteins. Notably, glycation of cortical bone from older donors led to much higher AGEs levels as compared to young donors. Such efficient in vitro glycation of older cortical bone could result from aging-related increase in porosity caused by the loss of mineral content. In addition, more pronounced glycation in vivo would be driven by elevated oxidation processes. Interestingly, the levels of PEN formation differed pronouncedly between glucosylation and ribosylation. Ribosylation generated very high levels of PEN (approx. 6- vs. 2.5-fold higher PEN level than in glucosylated samples). Kinetic studies of AGEs and PEN formation in human cortical and cancellous bone matrix confirmed higher accumulation of fluorescent crosslinks for ribosylation. Our results suggest that in vitro glycation of bone using glucose leads to the formation of lower levels of AGEs including PEN, whereas ribosylation appears to support a pathway toward PEN formation. Our studies may help to understand differences in the progression of bone pathologies related to protein glycation by different sugars, and raise awareness for excessive sugar supplementation in food and drinks.
Collapse
Affiliation(s)
- Grażyna E. Sroga
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Alankrita Siddula
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| |
Collapse
|
43
|
Dwyer JP, Greco BA, Umanath K, Packham D, Fox JW, Peterson R, Broome BR, Greene LE, Sika M, Lewis JB. Pyridoxamine dihydrochloride in diabetic nephropathy (PIONEER-CSG-17): lessons learned from a pilot study. Nephron Clin Pract 2014; 129:22-8. [PMID: 25532068 DOI: 10.1159/000369310] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 10/02/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Pyridoxamine dihydrochloride (Pyridorin™) blocks pathogenic oxidative pathways in the progression of diabetic nephropathy. The pyridoxamine pilot study was designed to test entry criteria and outcomes. Subjects had SCr 1.3-3.5 mg/dl, protein-to-creatinine ≥1,200 mg/g and used a surrogate outcome of ΔSCr over 52 weeks. Subjects had to be on a maximally tolerated dose of ACE/ARB for 3 months; stable other antihypertensive doses for 2 months; stable diuretic dose for 2 weeks, and BP ≤160/90 mm Hg; or enter a Pharmaco-Stabilization Phase (PSP). This pilot failed to detect an effect on ΔSCr in intent-to-treat analysis. METHODS We queried the locked clinical trial database for subgroups in which there was a treatment effect. RESULTS Subjects not requiring PSP and those with entry SCr <2.0 mg/dl had a treatment effect. Subjects entering PSP required more changes in antihypertensive medications and experienced larger ΔSCr over 52 weeks. PSP subjects with BP >140/90 mm Hg had no treatment effect, but those ≤140/90 mm Hg did. CONCLUSION Time required for acute effects of ACE/ARB to stabilize is unknown, but these data suggest >3 months. Thus, subjects in the pivotal trial must be on ACE/ARB for 6 months. Frequent antihypertensive adjustment could engender SCr changes unrelated to CKD progression. Thus, we will require subjects to have BP ≤150/90 mm Hg and on stable antihypertensives for 26 weeks, or ≤140/90 mm Hg and on stable antihypertensives for 13 weeks. Since ΔSCr over 52 weeks is limited as a surrogate outcome, the pivotal trial uses a time-to-event analysis of baseline SCr to at least a 50% increase in SCr or ESRD as the primary outcome. This substantial ΔSCr is protected from noise and is clinically relevant. The pyridoxamine pilot provided critical information to inform the design of PIONEER-CSG-17, which we conducted under the SPA agreement with FDA.
Collapse
|
44
|
Grove KJ, Voziyan PA, Spraggins JM, Wang S, Paueksakon P, Harris RC, Hudson BG, Caprioli RM. Diabetic nephropathy induces alterations in the glomerular and tubule lipid profiles. J Lipid Res 2014; 55:1375-85. [PMID: 24864273 DOI: 10.1194/jlr.m049189] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Indexed: 12/19/2022] Open
Abstract
Diabetic nephropathy (DN) is a major life-threatening complication of diabetes. Renal lesions affect glomeruli and tubules, but the pathogenesis is not completely understood. Phospholipids and glycolipids are molecules that carry out multiple cell functions in health and disease, and their role in DN pathogenesis is unknown. We employed high spatial resolution MALDI imaging MS to determine lipid changes in kidneys of eNOS(-/-) db/db mice, a robust model of DN. Phospholipid and glycolipid structures, localization patterns, and relative tissue levels were determined in individual renal glomeruli and tubules without disturbing tissue morphology. A significant increase in the levels of specific glomerular and tubular lipid species from four different classes, i.e., gangliosides, sulfoglycosphingolipids, lysophospholipids, and phosphatidylethanolamines, was detected in diabetic kidneys compared with nondiabetic controls. Inhibition of nonenzymatic oxidative and glycoxidative pathways attenuated the increase in lipid levels and ameliorated renal pathology, even though blood glucose levels remained unchanged. Our data demonstrate that the levels of specific phospho- and glycolipids in glomeruli and/or tubules are associated with diabetic renal pathology. We suggest that hyperglycemia-induced DN pathogenic mechanisms require intermediate oxidative steps that involve specific phospholipid and glycolipid species.
Collapse
Affiliation(s)
- Kerri J Grove
- Departments of Chemistry, Vanderbilt University Medical Center, Nashville, TN 37232 Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37232 Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Paul A Voziyan
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37232 Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Suwan Wang
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Paisit Paueksakon
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Raymond C Harris
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Billy G Hudson
- Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Richard M Caprioli
- Departments of Chemistry, Vanderbilt University Medical Center, Nashville, TN 37232 Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37232 Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
45
|
Tsekovska R, Boyanova M, Mironova R, Ivanov I. Impact of Glycation Inhibitors on the Biologic Activity of Recombinant Human Interferon-Gamma. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.5504/50yrtimb.2011.0031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
46
|
Inhibition of fluorescent advanced glycation end products (AGEs) of human serum albumin upon incubation with 3-β-hydroxybutyrate. Mol Biol Rep 2014; 41:3705-13. [DOI: 10.1007/s11033-014-3235-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 02/06/2014] [Indexed: 11/26/2022]
|
47
|
Elseweidy MM, Elswefy SE, Younis NN, Zaghloul MS. Pyridoxamine, an inhibitor of protein glycation, in relation to microalbuminuria and proinflammatory cytokines in experimental diabetic nephropathy. Exp Biol Med (Maywood) 2014; 238:881-8. [PMID: 23970406 DOI: 10.1177/1535370213494644] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the major complications that develop as consequence of chronic and uncontrolled hyperglycaemia. Hyperglycaemia initiates various processes, one of which is protein glycation, leading to the formation of advanced glycation end products. Alteration of intracellular signalling, gene expression, release of proinflammatory molecules and free radicals are examples of such changes and they contribute to the initiation of diabetic complications. In the current manuscript, we studied the effect of pyridoxamine (PM) on protein glycation, oxidative stress, interleukin-1α (IL-1α), IL-6, C-reactive protein (CRP), gene expression of tumour necrosis factor-α (TNF-α) and transforming growth factor-β1 (TGF-β1) in relation to microalbuminuria and kidney functions in a model of alloxan-induced diabetic rats. We have observed that onset of microalbuminuria has preceded the gradual increase of blood sugar level in diabetic rats. In diabetic rats, gene expression of TNF-α and TGF-β1 recorded a gradual increase and marked increase was observed after one and two weeks of alloxan administration, in comparison with normal rats. PM induced significant decrease in kidney malondialdehyde content and the gene expression of TNF-α and TGF-β1, in addition to levels of serum glucose, fructosamine, urea, creatinine, IL-1α, IL-6, CRP and urine microalbumin. Histopathological examination of kidney tissues showed certain improvements as compared with diabetic control. In conclusion, our results may provide a supporting evidence for the therapeutic benefit of PM in DN.
Collapse
|
48
|
Brodeur MR, Bouvet C, Bouchard S, Moreau S, Leblond J, deBlois D, Moreau P. Reduction of advanced-glycation end products levels and inhibition of RAGE signaling decreases rat vascular calcification induced by diabetes. PLoS One 2014; 9:e85922. [PMID: 24465790 PMCID: PMC3897559 DOI: 10.1371/journal.pone.0085922] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/03/2013] [Indexed: 11/18/2022] Open
Abstract
Advanced-glycation end products (AGEs) were recently implicated in vascular calcification, through a process mediated by RAGE (receptor for AGEs). Although a correlation between AGEs levels and vascular calcification was established, there is no evidence that reducing in vivo AGEs deposition or inhibiting AGEs-RAGE signaling pathways can decrease medial calcification. We evaluated the impact of inhibiting AGEs formation by pyridoxamine or elimination of AGEs by alagebrium on diabetic medial calcification. We also evaluated if the inhibition of AGEs-RAGE signaling pathways can prevent calcification. Rats were fed a high fat diet during 2 months before receiving a low dose of streptozotocin. Then, calcification was induced with warfarin. Pyridoxamine was administered at the beginning of warfarin treatment while alagebrium was administered 3 weeks after the beginning of warfarin treatment. Results demonstrate that AGEs inhibitors prevent the time-dependent accumulation of AGEs in femoral arteries of diabetic rats. This effect was accompanied by a reduced diabetes-accelerated calcification. Ex vivo experiments showed that N-methylpyridinium, an agonist of RAGE, induced calcification of diabetic femoral arteries, a process inhibited by antioxidants and different inhibitors of signaling pathways associated to RAGE activation. The physiological importance of oxidative stress was demonstrated by the reduction of femoral artery calcification in diabetic rats treated with apocynin, an inhibitor of reactive oxygen species production. We demonstrated that AGE inhibitors prevent or limit medial calcification. We also showed that diabetes-accelerated calcification is prevented by antioxidants. Thus, inhibiting the association of AGE-RAGE or the downstream signaling reduced medial calcification in diabetes.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Aorta/metabolism
- Aorta/pathology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Femoral Artery/drug effects
- Femoral Artery/pathology
- Femoral Artery/physiopathology
- Glycation End Products, Advanced/metabolism
- Hemodynamics/drug effects
- Male
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxidation-Reduction/drug effects
- Pyridinium Compounds
- Rats
- Rats, Wistar
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/metabolism
- Signal Transduction/drug effects
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/physiopathology
Collapse
Affiliation(s)
| | - Céline Bouvet
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Sonia Bouchard
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Simon Moreau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Jeanne Leblond
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Denis deBlois
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Moreau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
49
|
Bohlooli M, Moosavi-Movahedi A, F. Taghavi, Maghami P, Saboury A, Moosavi-Movahedi Z, Farhadi M, Hong J, Sheibani N, Habibi-Rezaei M. Investigation of thermal reversibility and stability of glycated human serum albumin. Int J Biol Macromol 2013; 62:358-64. [DOI: 10.1016/j.ijbiomac.2013.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 09/17/2013] [Accepted: 09/17/2013] [Indexed: 12/27/2022]
|
50
|
Inhibitory effect of metformin and pyridoxamine in the formation of early, intermediate and advanced glycation end-products. PLoS One 2013; 8:e72128. [PMID: 24023728 PMCID: PMC3762829 DOI: 10.1371/journal.pone.0072128] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 07/05/2013] [Indexed: 11/19/2022] Open
Abstract
Background Non-enzymatic glycation is the addition of free carbonyl group of reducing sugar to the free amino groups of proteins, resulting in the formation of a Schiff base and an Amadori product. Dihydroxyacetone (DHA) is one of the carbonyl species which reacts rapidly with the free amino groups of proteins to form advanced glycation end products (AGEs). The highly reactive dihydroxyacetone phosphate is a derivative of dihydroxyacetone (DHA), and a product of glycolysis, having potential glycating effects to form AGEs. The formation of AGEs results in the generation of free radicals which play an important role in the pathophysiology of aging and diabetic complications. While the formation of DHA-AGEs has been demonstrated previously, no extensive studies have been performed to assess the inhibition of AGE inhibitors at all the three stages of glycation (early, intermediate and late) using metformin (MF) and pyridoxamine (PM) as a novel inhibitor. Methodology/Principal Findings In this study we report glycation of human serum albumin (HSA) & its characterization by various spectroscopic techniques. Furthermore, inhibition of glycation products at all the stages of glycation was also studied. Spectroscopic analysis suggests structural perturbations in the HSA as a result of modification which might be due to generation of free radicals and formation of AGEs. Conclusion The inhibition in the formation of glycation reaction reveals that Pyridoxamine is a better antiglycating agent than Metformin at all stages of the glycation (early, intermediate and late stages).
Collapse
|