1
|
Rufin M, Nalbach M, Rakuš M, Fuchs M, Poik M, Schitter G, Thurner PJ, Andriotis OG. Methylglyoxal alters collagen fibril nanostiffness and surface potential. Acta Biomater 2024; 189:208-216. [PMID: 39218277 DOI: 10.1016/j.actbio.2024.08.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Collagen fibrils are fundamental to the mechanical strength and function of biological tissues. However, they are susceptible to changes from non-enzymatic glycation, resulting in the formation of advanced glycation end-products (AGEs) that are not reversible. AGEs accumulate with aging and disease and can adversely impact tissue mechanics and cell-ECM interactions. AGE-crosslinks have been related, on the one hand, to dysregulation of collagen fibril stiffness and damage and, on the other hand, to altered collagen net surface charge as well as impaired cell recognition sites. While prior studies using Kelvin probe force microscopy (KPFM) have shown the effect glycation has on collagen fibril surface potential (i.e., net charge), the combined effect on individual and isolated collagen fibril mechanics, hydration, and surface potential has not been documented. Here, we explore how methylglyoxal (MGO) treatment affects the mechanics and surface potential of individual and isolated collagen fibrils by utilizing atomic force microscopy (AFM) nanoindentation and KPFM. Our results reveal that MGO treatment significantly increases nanostiffness, alters surface potential, and modifies hydration characteristics at the collagen fibril level. These findings underscore the critical impact of AGEs on collagen fibril physicochemical properties, offering insights into pathophysiological mechanical and biochemical alterations with implications for cell mechanotransduction during aging and in diabetes. STATEMENT OF SIGNIFICANCE: Collagen fibrils are susceptible to glycation, the irreversible reaction of amino acids with sugars. Glycation affects the mechanical properties and surface chemistry of collagen fibrils with adverse alterations in biological tissue mechanics and cell-ECM interactions. Current research on glycation, at the level of individual collagen fibrils, is sparse and has focused either on collagen fibril mechanics, with contradicting evidence, or surface potential. Here, we utilized a multimodal approach combining Kelvin probe force (KPFM) and atomic force microscopy (AFM) to examine how methylglyoxal glycation induces structural, mechanical, and surface potential changes on the same individual and isolated collagen fibrils. This approach helps inform structure-function relationships at the level of individual collagen fibrils.
Collapse
Affiliation(s)
- Manuel Rufin
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Mathis Nalbach
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Maja Rakuš
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Magdalena Fuchs
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Mathias Poik
- Automation and Control Institute (ACIN), TU Wien, Gusshausstrasse 27-29, A-1040 Vienna, Austria
| | - Georg Schitter
- Automation and Control Institute (ACIN), TU Wien, Gusshausstrasse 27-29, A-1040 Vienna, Austria
| | - Philipp J Thurner
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Orestis G Andriotis
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria.
| |
Collapse
|
2
|
Ewald CY, Nyström A. Mechanotransduction through hemidesmosomes during aging and longevity. J Cell Sci 2023; 136:jcs260987. [PMID: 37522320 DOI: 10.1242/jcs.260987] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
Hemidesmosomes are structural protein complexes localized at the interface of tissues with high mechanical demand and shear forces. Beyond tissue anchoring, hemidesmosomes have emerged as force-modulating structures important for translating mechanical cues into biochemical and transcriptional adaptation (i.e. mechanotransduction) across tissues. Here, we discuss the recent insights into the roles of hemidesmosomes in age-related tissue regeneration and aging in C. elegans, mice and humans. We highlight the emerging concept of preserved dynamic mechanoregulation of hemidesmosomes in tissue maintenance and healthy aging.
Collapse
Affiliation(s)
- Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zürich, Schwerzenbach CH-8603, Switzerland
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg DE-79104, Germany
- Freiburg Institute for Advanced Studies (FRIAS), Albertstraße 19, Freiburg im Breisgau DE-79104, Germany
| |
Collapse
|
3
|
Genova VM, Gambero A, de Souza Freitas Campos P, Macedo GA. Polyphenolic Compounds Mechanisms as Inhibitors of Advanced Glycation End Products and Their Relationship to Health and Disease. MOLECULAR MECHANISMS OF FUNCTIONAL FOOD 2022:1-27. [DOI: 10.1002/9781119804055.ch1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Costello L, Dicolandrea T, Tasseff R, Isfort R, Bascom C, von Zglinicki T, Przyborski S. Tissue engineering strategies to bioengineer the ageing skin phenotype in vitro. Aging Cell 2022; 21:e13550. [PMID: 35037366 PMCID: PMC8844123 DOI: 10.1111/acel.13550] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 11/29/2022] Open
Abstract
Human skin ageing is a complex and heterogeneous process, which is influenced by genetically determined intrinsic factors and accelerated by cumulative exposure to extrinsic stressors. In the current world ageing demographic, there is a requirement for a bioengineered ageing skin model, to further the understanding of the intricate molecular mechanisms of skin ageing, and provide a distinct and biologically relevant platform for testing actives and formulations. There have been many recent advances in the development of skin models that recapitulate aspects of the ageing phenotype in vitro. This review encompasses the features of skin ageing, the molecular mechanisms that drive the ageing phenotype, and tissue engineering strategies that have been utilised to bioengineer ageing skin in vitro.
Collapse
Affiliation(s)
| | | | - Ryan Tasseff
- Procter and GambleMason Business CenterCincinnatiOhioUSA
| | - Robert Isfort
- Procter and GambleMason Business CenterCincinnatiOhioUSA
| | - Charlie Bascom
- Procter and GambleMason Business CenterCincinnatiOhioUSA
| | - Thomas von Zglinicki
- Institute for Cell and Molecular SciencesNewcastle UniversityNewcastle Upon TyneUK
| | - Stefan Przyborski
- Department of BiosciencesDurham UniversityDurhamUK
- Reprocell EuropeGlasgow, DurhamUK
| |
Collapse
|
5
|
André A, Touré AK, Stien D, Eparvier V. 2,5-diketopiperazines mitigate the amount of advanced glycation end products accumulated with age in human dermal fibroblasts. Int J Cosmet Sci 2020; 42:596-604. [PMID: 32767373 DOI: 10.1111/ics.12655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 07/29/2020] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Glycation is a common non-enzymatic reaction between proteins and sugars, resulting in the formation of advanced glycation end products (AGEs) in the human body. As can be seen in diabetic patients, the accumulation of AGEs in the skin has aesthetic consequences (wrinkles, brown spots and yellowish complexion). Therefore, the objective of this work was to find compounds isolated from natural sources that could eliminate the final AGEs accumulated in the skin with ageing. METHODS AND RESULTS A preliminary screening performed on a bank of microbial extracts and pure compounds showed that 2,5-Diketopiperazines (DKPs), as well as the extract of Sphingobacterium sp (SNB-CN13), reduced the presence of AGEs in fibroblasts by -28% and -23%, respectively. In this article, we present the dereplication approach used to reveal the presence of 26 different DKPs in the crude extract of Sphingobacterium sp. Bioguided fractionation has led to the isolation of 12 of them, whose identity has been confirmed by HRMS and NMR. A green synthesis approach has been developed to synthesize 3 symmetrical DKPs. The biological activity of all DKPs was evaluated by the development of an in vitro test using immunocytochemistry to reveal the presence of AGE carboxymethyl-lysine in human dermal fibroblasts. CONCLUSION Our work shows for the first time that DKPs decrease the amount of carboxymethyl-lysine AGE in elderly human dermal fibroblasts grown in vitro. Therefore, diketopiperazines can be considered as compounds of interest for dermatological and cosmetic applications with an anti-ageing aim.
Collapse
Affiliation(s)
- A André
- CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, Université Paris-Saclay, 1 avenue de la Terrasse, Gif-sur-Yvette, 91198, France.,Laboratoire Shigeta, 62 boulevard Davout, Paris, 75020, France
| | - A K Touré
- Laboratoire Shigeta, 62 boulevard Davout, Paris, 75020, France
| | - D Stien
- Laboratoire de Biodiversité et Biotechnologies Microbiennes, Sorbonne Université, CNRS, USR 3579, Banyuls-sur-mer, 66650, France
| | - V Eparvier
- Laboratoire Shigeta, 62 boulevard Davout, Paris, 75020, France
| |
Collapse
|
6
|
Ravichandran G, Lakshmanan DK, Raju K, Elangovan A, Nambirajan G, Devanesan AA, Thilagar S. Food advanced glycation end products as potential endocrine disruptors: An emerging threat to contemporary and future generation. ENVIRONMENT INTERNATIONAL 2019; 123:486-500. [PMID: 30622074 DOI: 10.1016/j.envint.2018.12.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 06/09/2023]
Abstract
Mankind exposure to chemicals in the past century has increased dramatically throughout environment. There is no question that chemicals interfere with the physiology of biological system. Abundance of chemicals is documented to be detrimental to human and wildlife. The mammalian endocrine system is comprised of many interacting tissues mediate themselves through hormones that are essential for metabolism, growth and development. Humans secrete over fifty different hormones to orchestrate major physiological functions however; these vital functions can be intervened by huge number of internal and external chemical stressors that are identified as endocrine disruptors. Advanced glycation end products (AGEs), familiarly known as Maillard products, formed through non-enzymatic glycation whose production is augmented on aging as well as environmental stressors. Processed foods have become very popular today due to their taste, convenience, and inexpensiveness. Manufacture of these day-to-day foods involves extreme temperatures on processing results in the formation of AGEs could independently promote oxidative stress, aging, diabetes, cancer, degenerative diseases, more fascinatingly hormonal disruption is the subject of interest of this review. Based on some substantial observations documented till time, we discuss the emergence of dietary AGEs as potential endocrine disruptors by emphasizing their occurrence, mechanisms and participation in endocrine interruption. Both economically and in terms of human life, AGEs may represent an enormous cost for the future society. Therefore, by explicating their novel role in endocrine diseases, the review strives to make an impact on AGEs and their exposure among public as well as scientific communities.
Collapse
Affiliation(s)
- Guna Ravichandran
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Dinesh Kumar Lakshmanan
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Karthik Raju
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Abbirami Elangovan
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Gayathri Nambirajan
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Arul Ananth Devanesan
- Department of Food Quality and Safety, Gilat Research Center, Agricultural Research Organization, M.P. Negev 85280, Israel
| | - Sivasudha Thilagar
- Department of Environmental Biotechnology, School of Environmental Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu, India.
| |
Collapse
|
7
|
Ferreira JSSP, Panighel JP, Silva ÉQ, Monteiro RL, Cruvinel Júnior RH, Sacco ICN. Foot function and strength of patients with diabetes grouped by ulcer risk classification (IWGDF). Diabetol Metab Syndr 2019; 11:89. [PMID: 31695753 PMCID: PMC6822353 DOI: 10.1186/s13098-019-0487-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/22/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The stratification system from the International Working Group on the Diabetic Foot (IWGDF) was used to classify the participants as to the ulcer risk. However, it is not yet known what the classification groups' individual deficits are regarding sensitivity, function, and musculoskeletal properties and mechanics. This makes it difficult to design proper ulcer prevention strategies for patients. Thus, this study aimed to investigate the foot function, foot strength and health of people with diabetes mellitus (DM)-with or without DPN-while considering the different ulcer risk classifications determined by the IWGDF. METHODS The subject pool comprised 72 people with DM, with and without DPN. The patients were divided into three groups: Group 0 (G0), which comprised diabetic patients without DPN; Group 1 (G1), which comprised patients with DPN; and Group 2 (G2), which comprised patients with DPN who had foot deformities. The health and foot function of the subjects' feet were assessed using a foot health status questionnaire (FHSQ-BR) that investigated four domains: foot pain, foot function, footwear, and general foot health. The patients' foot strength was evaluated using the maximum force under each subject's hallux and toes on a pressure platform (emed q-100, Novel, Munich, Germany). RESULTS Moderate differences were found between G0 and G1 and G2 for the foot pain, foot function, general foot health, and footwear. There was also a small but significant difference between G0 and G2 in regards to hallux strength. CONCLUSION Foot health, foot function and strength levels of people with DM and DPN classified by the ulcer risk are different and this must be taken into account when evaluating and developing treatment strategies for these patients.
Collapse
Affiliation(s)
- Jane S. S. P. Ferreira
- Department of Physical Therapy, Speech, and Occupational Therapy, School of Medicine, University of São Paulo, Rua Cipotânea, 51-Cidade Universitária, São Paulo, 05360-160 Brazil
| | - João P. Panighel
- Department of Physical Therapy, Speech, and Occupational Therapy, School of Medicine, University of São Paulo, Rua Cipotânea, 51-Cidade Universitária, São Paulo, 05360-160 Brazil
| | - Érica Q. Silva
- Department of Physical Therapy, Speech, and Occupational Therapy, School of Medicine, University of São Paulo, Rua Cipotânea, 51-Cidade Universitária, São Paulo, 05360-160 Brazil
| | - Renan L. Monteiro
- Department of Physical Therapy, Speech, and Occupational Therapy, School of Medicine, University of São Paulo, Rua Cipotânea, 51-Cidade Universitária, São Paulo, 05360-160 Brazil
- Departamento de Ciências da Saúde, Universidade Federal do Amapá, Macapá, Amapá Brazil
| | - Ronaldo H. Cruvinel Júnior
- Department of Physical Therapy, Speech, and Occupational Therapy, School of Medicine, University of São Paulo, Rua Cipotânea, 51-Cidade Universitária, São Paulo, 05360-160 Brazil
| | - Isabel C. N. Sacco
- Department of Physical Therapy, Speech, and Occupational Therapy, School of Medicine, University of São Paulo, Rua Cipotânea, 51-Cidade Universitária, São Paulo, 05360-160 Brazil
| |
Collapse
|
8
|
Sorushanova A, Delgado LM, Wu Z, Shologu N, Kshirsagar A, Raghunath R, Mullen AM, Bayon Y, Pandit A, Raghunath M, Zeugolis DI. The Collagen Suprafamily: From Biosynthesis to Advanced Biomaterial Development. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1801651. [PMID: 30126066 DOI: 10.1002/adma.201801651] [Citation(s) in RCA: 599] [Impact Index Per Article: 99.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/03/2018] [Indexed: 05/20/2023]
Abstract
Collagen is the oldest and most abundant extracellular matrix protein that has found many applications in food, cosmetic, pharmaceutical, and biomedical industries. First, an overview of the family of collagens and their respective structures, conformation, and biosynthesis is provided. The advances and shortfalls of various collagen preparations (e.g., mammalian/marine extracted collagen, cell-produced collagens, recombinant collagens, and collagen-like peptides) and crosslinking technologies (e.g., chemical, physical, and biological) are then critically discussed. Subsequently, an array of structural, thermal, mechanical, biochemical, and biological assays is examined, which are developed to analyze and characterize collagenous structures. Lastly, a comprehensive review is provided on how advances in engineering, chemistry, and biology have enabled the development of bioactive, 3D structures (e.g., tissue grafts, biomaterials, cell-assembled tissue equivalents) that closely imitate native supramolecular assemblies and have the capacity to deliver in a localized and sustained manner viable cell populations and/or bioactive/therapeutic molecules. Clearly, collagens have a long history in both evolution and biotechnology and continue to offer both challenges and exciting opportunities in regenerative medicine as nature's biomaterial of choice.
Collapse
Affiliation(s)
- Anna Sorushanova
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Luis M Delgado
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Zhuning Wu
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Naledi Shologu
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Aniket Kshirsagar
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rufus Raghunath
- Centre for Cell Biology and Tissue Engineering, Competence Centre Tissue Engineering for Drug Development (TEDD), Department Life Sciences and Facility Management, Institute for Chemistry and Biotechnology (ICBT), Zürich University of Applied Sciences, Wädenswil, Switzerland
| | | | - Yves Bayon
- Sofradim Production-A Medtronic Company, Trevoux, France
| | - Abhay Pandit
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Centre for Cell Biology and Tissue Engineering, Competence Centre Tissue Engineering for Drug Development (TEDD), Department Life Sciences and Facility Management, Institute for Chemistry and Biotechnology (ICBT), Zürich University of Applied Sciences, Wädenswil, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| |
Collapse
|
9
|
Ngai D, Lino M, Bendeck MP. Cell-Matrix Interactions and Matricrine Signaling in the Pathogenesis of Vascular Calcification. Front Cardiovasc Med 2018; 5:174. [PMID: 30581820 PMCID: PMC6292870 DOI: 10.3389/fcvm.2018.00174] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification is a complex pathological process occurring in patients with atherosclerosis, type 2 diabetes, and chronic kidney disease. The extracellular matrix, via matricrine-receptor signaling plays important roles in the pathogenesis of calcification. Calcification is mediated by osteochondrocytic-like cells that arise from transdifferentiating vascular smooth muscle cells. Recent advances in our understanding of the plasticity of vascular smooth muscle cell and other cells of mesenchymal origin have furthered our understanding of how these cells transdifferentiate into osteochondrocytic-like cells in response to environmental cues. In the present review, we examine the role of the extracellular matrix in the regulation of cell behavior and differentiation in the context of vascular calcification. In pathological calcification, the extracellular matrix not only provides a scaffold for mineral deposition, but also acts as an active signaling entity. In recent years, extracellular matrix components have been shown to influence cellular signaling through matrix receptors such as the discoidin domain receptor family, integrins, and elastin receptors, all of which can modulate osteochondrocytic differentiation and calcification. Changes in extracellular matrix stiffness and composition are detected by these receptors which in turn modulate downstream signaling pathways and cytoskeletal dynamics, which are critical to osteogenic differentiation. This review will focus on recent literature that highlights the role of cell-matrix interactions and how they influence cellular behavior, and osteochondrocytic transdifferentiation in the pathogenesis of cardiovascular calcification.
Collapse
Affiliation(s)
- David Ngai
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Marsel Lino
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
André A, Wdzieczak-Bakala J, Touré AK, Stien D, Eparvier V. A method to quantify intracellular glycation in dermal fibroblasts using liquid chromatography coupled to fluorescence detection – Application to the selection of deglycation compounds of dermatological interest. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1100-1101:100-105. [DOI: 10.1016/j.jchromb.2018.09.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 10/28/2022]
|
11
|
Fournet M, Bonté F, Desmoulière A. Glycation Damage: A Possible Hub for Major Pathophysiological Disorders and Aging. Aging Dis 2018; 9:880-900. [PMID: 30271665 PMCID: PMC6147582 DOI: 10.14336/ad.2017.1121] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/21/2017] [Indexed: 12/25/2022] Open
Abstract
Glycation is both a physiological and pathological process which mainly affects proteins, nucleic acids and lipids. Exogenous and endogenous glycation produces deleterious reactions that take place principally in the extracellular matrix environment or within the cell cytosol and organelles. Advanced glycation end product (AGE) formation begins by the non-enzymatic glycation of free amino groups by sugars and aldehydes which leads to a succession of rearrangements of intermediate compounds and ultimately to irreversibly bound products known as AGEs. Epigenetic factors, oxidative stress, UV and nutrition are important causes of the accumulation of chemically and structurally different AGEs with various biological reactivities. Cross-linked proteins, deriving from the glycation process, present both an altered structure and function. Nucleotides and lipids are particularly vulnerable targets which can in turn favor DNA mutation or a decrease in cell membrane integrity and associated biological pathways respectively. In mitochondria, the consequences of glycation can alter bioenergy production. Under physiological conditions, anti-glycation defenses are sufficient, with proteasomes preventing accumulation of glycated proteins, while lipid turnover clears glycated products and nucleotide excision repair removes glycated nucleotides. If this does not occur, glycation damage accumulates, and pathologies may develop. Glycation-induced biological products are known to be mainly associated with aging, neurodegenerative disorders, diabetes and its complications, atherosclerosis, renal failure, immunological changes, retinopathy, skin photoaging, osteoporosis, and progression of some tumors.
Collapse
Affiliation(s)
- Maxime Fournet
- 1University of Limoges, Faculty of Pharmacy, Department of Physiology, EA 6309, F-87025 Limoges, France
| | | | - Alexis Desmoulière
- 3University of Limoges, Faculty of Pharmacy, Department of Physiology, EA 6309, F-87025 Limoges, France
| |
Collapse
|
12
|
Limonene: Aroma of innovation in health and disease. Chem Biol Interact 2018; 283:97-106. [PMID: 29427589 DOI: 10.1016/j.cbi.2018.02.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
Abstract
Natural products obtained in dietary components may aid the prevention and treatment of a variety of diseases. Reports in the scientific literature have demonstrated that the consumption of terpenes is a successful alternative in the treatment of several diseases, triggering beneficial biological effects in clinical and preclinical studies. The monoterpene limonene is largely used in alimentary items, cleaning products, and it is one of the most frequent fragrances used in cosmetics formulation. The therapeutic effects of limonene have been extensively studied, proving anti-inflammatory, antioxidant, antinociceptive, anticancer, antidiabetic, antihyperalgesic, antiviral, and gastroprotective effects, among other beneficial effects in health. In this review, we collected, presented, and analyzed evidence from the scientific literature regarding the usage of limonene and its activities and underlying mechanisms involved in combating diseases. The highlighting of limonene applications could develop a useful targeting of innovative research in this field as well as the development of a limonene-based phytomedicine which could be used in a variety of conditions of health and disease.
Collapse
|
13
|
Chen C, Gong W, Li C, Xiong F, Wang S, Huang J, Wang Y, Chen Z, Chen Q, Liu P, Lan T, Huang H. Sphingosine kinase 1 mediates AGEs-induced fibronectin upregulation in diabetic nephropathy. Oncotarget 2017; 8:78660-78676. [PMID: 29108256 PMCID: PMC5667989 DOI: 10.18632/oncotarget.20205] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Activation of sphingosine kinase 1 (SphK1) signaling pathway mediates fibronectin (FN) upregulation in glomerular mesangial cells (GMCs) under high glucose (HG) condition. However, the roles of SphK1 in advanced glycation end products (AGEs)-induced DN have not been elucidated. Here we show that AGEs upregulated FN and SphK1 and SphK1 activity. Inhibition of SphK1 signaling attenuated AGEs-induced FN synthesis in GMCs. Inhibition of AGE receptor (RAGE) signaling reduced the upregulation of FN and SphK1 and SphK1 activity in GMCs induced by AGEs. Treatment of aminoguanidine ameliorates the renal injury and fibrosis in STZ-induced diabetic mice and attenuated SphK1 expression and activity in diabetic mouse kidneys. The renal injury and fibrosis in diabetic SphK1-/- mice was significantly attenuated than WT mice. Furthermore, AGEs upregulated SphK1 by reducing its degradation and prolonging its half-life. CONCLUSION SphK1 mediates AGEs-induced FN synthesis in GMCs and diabetic mice under hyperglycemic condition.
Collapse
Affiliation(s)
- Cheng Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National & Local United Engineering Lab of Drug Screening and Evaluation, Guangzhou 510006, China
| | - Wenyan Gong
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Changzheng Li
- Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Fengxiao Xiong
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shaogui Wang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junying Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yu Wang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiquan Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiuhong Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Peiqing Liu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tian Lan
- Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National & Local United Engineering Lab of Drug Screening and Evaluation, Guangzhou 510006, China
| |
Collapse
|
14
|
Rabie E, Serem JC, Oberholzer HM, Gaspar ARM, Bester MJ. How methylglyoxal kills bacteria: An ultrastructural study. Ultrastruct Pathol 2016; 40:107-11. [PMID: 26986806 DOI: 10.3109/01913123.2016.1154914] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Antibacterial activity of honey is due to the presence of methylglyoxal (MGO), H2O2, bee defensin as well as polyphenols. High MGO levels in manuka honey are the main source of antibacterial activity. Manuka honey has been reported to reduce the swarming and swimming motility of Pseudomonas aeruginosa due to de-flagellation. Due to the complexity of honey it is unknown if this effect is directly due to MGO. In this ultrastructural investigation the effects of MGO on the morphology of bacteria and specifically the structure of fimbriae and flagella were investigated. MGO effectively inhibited Gram positive (Bacillus subtilis; MIC 0.8 mM and Staphylococcus aureus; MIC 1.2 mM) and Gram negative (P. aeruginosa; MIC 1.0 mM and Escherichia coli; MIC 1.2 mM) bacteria growth. The ultrastructural effects of 0.5, 1.0 and 2 mM MGO on B. substilis and E. coli morphology was then evaluated. At 0.5 mM MGO, bacteria structure was unaltered. For both bacteria at 1 mM MGO fewer fimbriae were present and the flagella were less or absent. Identified structures appeared stunted and fragile. At 2 mM MGO fimbriae and flagella were absent while the bacteria were rounded with shrinkage and loss of membrane integrity. Antibacterial MGO causes alterations in the structure of bacterial fimbriae and flagella which would limit bacteria adherence and motility.
Collapse
Affiliation(s)
- Erika Rabie
- a Department of Anatomy, Faculty of Health Sciences , University of Pretoria , Arcadia , South Africa
| | - June Cheptoo Serem
- a Department of Anatomy, Faculty of Health Sciences , University of Pretoria , Arcadia , South Africa
| | | | - Anabella Regina Marques Gaspar
- b Department of Biochemistry, Faculty of Natural and Agricultural Sciences , University of Pretoria , Arcadia , South Africa
| | - Megan Jean Bester
- a Department of Anatomy, Faculty of Health Sciences , University of Pretoria , Arcadia , South Africa
| |
Collapse
|
15
|
Monnier VM, Sun W, Gao X, Sell DR, Cleary PA, Lachin JM, Genuth S. Skin collagen advanced glycation endproducts (AGEs) and the long-term progression of sub-clinical cardiovascular disease in type 1 diabetes. Cardiovasc Diabetol 2015; 14:118. [PMID: 26341632 PMCID: PMC4560872 DOI: 10.1186/s12933-015-0266-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/31/2015] [Indexed: 11/10/2022] Open
Abstract
Background We recently reported strong associations between eight skin collagen AGEs and two solubility markers from skin biopsies obtained at DCCT study closeout and the long-term progression of microvascular disease in EDIC, despite adjustment for mean glycemia. Herein we investigated the hypothesis that some of these AGEs (fluorescence to be reported elsewhere) correlate with long-term subclinical cardiovascular disease (CVD) measurements, i.e. coronary artery calcium score (CAC) at EDIC year 7–9 (n = 187), change of carotid intima-media thickness (IMT) from EDIC year 1 to year 6 and 12 (n = 127), and cardiac MRI outcomes at EDIC year 15–16 (n = 142). Methods Skin collagen AGE measurements obtained from stored specimens were related to clinical data from the DCCT/EDIC using Spearman correlations and multivariable logistic regression analyses. Results Spearman correlations showed furosine (early glycation) was associated with future mean CAC (p < 0.05) and CAC >0 (p = 0.39), but not with CAC score <100 vs. >100. Glucosepane and pentosidine crosslinks, methylglyoxal hydroimidazolones (MG-H1) and pepsin solubility (inversely) correlated with IMT change from year 1 to 6(all P < 0.05). Left ventricular (LV) mass (cMRI) correlated with MG-H1, and inversely with pepsin solubility (both p < 0.05), while the ratio LV mass/end diastolic volume correlated with furosine and MG-H1 (both p < 0.05), and highly with CML (p < 0.01). In multivariate analysis only furosine (p = 0.01) was associated with CAC. In contrast IMT was inversely associated with lower collagen pepsin solubility and positively with glucosepane, Conclusions In type 1 diabetes, multiple AGEs are associated with IMT progression in spite of adjustment for A1c implying a likely participatory role of glycation and AGE mediated crosslinking on matrix accumulation in coronary arteries. This may also apply to functional cardiac MRI outcomes, especially left ventricular mass. In contrast, early glycation measured by furosine, but not AGEs, was associated with CAC score, implying hyperglycemia as a risk factor in calcium deposition perhaps via processes independent of glycation. Trial registration: Registered at Clinical trial reg. nos. NCT00360815 and NCT00360893, http://www.clinicaltrials.gov
Collapse
Affiliation(s)
- Vincent M Monnier
- Departments of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA. .,Departments of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Wanjie Sun
- The Biostatistics Center, The George Washington University, Rockville, MD, USA.
| | - Xiaoyu Gao
- The Biostatistics Center, The George Washington University, Rockville, MD, USA.
| | - David R Sell
- Departments of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Patricia A Cleary
- The Biostatistics Center, The George Washington University, Rockville, MD, USA.
| | - John M Lachin
- The Biostatistics Center, The George Washington University, Rockville, MD, USA.
| | - Saul Genuth
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | | |
Collapse
|
16
|
Yu S, Zhang W, Liu W, Zhu W, Guo R, Wang Y, Zhang D, Wang J. The inhibitory effect of selenium nanoparticles on protein glycation in vitro. NANOTECHNOLOGY 2015; 26:145703. [PMID: 25785463 DOI: 10.1088/0957-4484/26/14/145703] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Selenium nanoparticles (Se NPs) possess well-known excellent biological activities and low toxicity, and have been employed for numerous applications except as inhibitors to protein glycation. Herein, the present study is carried out to investigate the inhibitory effect of Se NPs on protein glycation in a bovine serum albumin (BSA)/glucose system. By measuring the amount of glucose covalently bound onto BSA, the formation of fructosamine and fluorescent products, it is found that Se NPs can hinder the development of protein glycation in a dose-dependent but time-independent manner under the selected reaction conditions (55 °C, 40 h). And after comparing the increase of inhibitory rate in different stages, it is observed that Se NPs show the greatest inhibitory effect in the early stage, then in the advanced stage, but no effect in the intermediate stage. Fourier transform infrared spectroscopy characterization of Se NPs collected after glycation and determination of ·OH influence and glyoxal formation show that the mechanism for the inhibitory efficacy of Se NPs is related to their strong competitive activity against available amino groups in proteins, their great scavenging activity on reactive oxygen species and their inhibitory effect on α-dicarbonyl compounds' formation. In addition, it is proved that Se NPs protect proteins from structural modifications in the system and they do not exhibit significant cytotoxicity towards BV-2 and BRL-3A cells at low concentrations (10 and 50 μg mL(-1)). Consequently, Se NPs may be suitable for further in vivo studies as novel anti-glycation agents.
Collapse
Affiliation(s)
- Shaoxuan Yu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Smith LE, White MY. The role of post-translational modifications in acute and chronic cardiovascular disease. Proteomics Clin Appl 2015; 8:506-21. [PMID: 24961403 DOI: 10.1002/prca.201400052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 05/27/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease (CVD) in one of the leading causes of mortality and morbidity worldwide, accounting for both primary diseases of the heart and vasculature and arising as a co-morbidity with numerous pathologies, including type 2 diabetes mellitus (T2DM). There has been significant emphasis on the role of the genome in CVD, aiding in the definition of 'at-risk' patients. The extent of disease penetrance however, can be influenced by environmental factors that are not detectable by investigating the genome alone. By targeting the transcriptome in response to CVD, the interplay between genome and environment is more apparent, however this implies the level of protein expression without reference to proteolytic turnover, or potentially more importantly, without defining the role of PTMs in the development of disease. Here, we discuss the role of both brief and irreversible PTMs in the setting of myocardial ischemia/reperfusion injury. Key proteins involved in calcium regulation have been observed as differentially modified by phosphorylation/O-GlcNAcylation or phosphorylation/redox modifications, with the level of interplay dependent on the physiological or pathophysiological state. The ability to modify crucial sites to produce the desired functional output is modulated by the presence of other PTMs as exemplified in the T2DM heart, where hyperglycemia results in aberrant O-GlcNAcylation and advanced glycation end products. By using the signalling events predicted to be critical to post-conditioning, an intervention with great promise for the cardioprotection of the ischemia/reperfusion injured heart, as an example, we discuss the level of PTMs and their interplay. The inability of post-conditioning to protect the diabetic heart may be regulated by aberrant PTMs influencing those sites necessary for protection.
Collapse
Affiliation(s)
- Lauren E Smith
- Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
18
|
Bodiga VL, Eda SR, Bodiga S. Advanced glycation end products: role in pathology of diabetic cardiomyopathy. Heart Fail Rev 2014; 19:49-63. [PMID: 23404649 DOI: 10.1007/s10741-013-9374-y] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence demonstrates that advanced glycation end products (AGEs) play a pivotal role in the development and progression of diabetic heart failure, although there are numerous other factors that mediate the disease response. AGEs are generated intra- and extracellularly as a result of chronic hyperglycemia. Then, following the interaction with receptors for advanced glycation end products (RAGEs), a series of events leading to vascular and myocardial damage are elicited and sustained, which include oxidative stress, increased inflammation, and enhanced extracellular matrix accumulation resulting in diastolic and systolic dysfunction. Whereas targeting glycemic control and treating additional risk factors, such as obesity, dyslipidemia, and hypertension, are mandatory to reduce chronic complications and prolong life expectancy in diabetic patients, drug therapy tailored to reducing the deleterious effects of the AGE-RAGE interactions is being actively investigated and showing signs of promise in treating diabetic cardiomyopathy and associated heart failure. This review shall discuss the formation of AGEs in diabetic heart tissue, potential targets of glycation in the myocardium, and underlying mechanisms that lead to diabetic cardiomyopathy and heart failure along with the use of AGE inhibitors and breakers in mitigating myocardial injury.
Collapse
Affiliation(s)
- Vijaya Lakshmi Bodiga
- Department of Biotechnology, Krishna University, Machilipatnam, Andhra Pradesh, India
| | | | | |
Collapse
|
19
|
Boonkaew B, Tompkins K, Manokawinchoke J, Pavasant P, Supaphol P. Characterization and cytological effects of a novel glycated gelatine substrate. Biomed Mater 2014; 9:025001. [DOI: 10.1088/1748-6041/9/2/025001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
20
|
Joglekar MM, Panaskar SN, Arvindekar AU. Inhibition of advanced glycation end product formation by cymene – A common food constituent. J Funct Foods 2014. [DOI: 10.1016/j.jff.2013.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
21
|
|
22
|
Pataridis S, Štastná Z, Sedláková P, Mikšík I. Monotopic modifications derived from in vitro glycation of albumin with ribose. Electrophoresis 2013; 34:1757-63. [DOI: 10.1002/elps.201300014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/14/2013] [Accepted: 01/18/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Statis Pataridis
- Institute of Physiology; Academy of Sciences of the Czech Republic v.v.i; Prague; Czech Republic
| | | | - Pavla Sedláková
- Institute of Physiology; Academy of Sciences of the Czech Republic v.v.i; Prague; Czech Republic
| | - Ivan Mikšík
- Institute of Physiology; Academy of Sciences of the Czech Republic v.v.i; Prague; Czech Republic
| |
Collapse
|
23
|
Höhn A, König J, Grune T. Protein oxidation in aging and the removal of oxidized proteins. J Proteomics 2013; 92:132-59. [PMID: 23333925 DOI: 10.1016/j.jprot.2013.01.004] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/08/2013] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are generated constantly within cells at low concentrations even under physiological conditions. During aging the levels of ROS can increase due to a limited capacity of antioxidant systems and repair mechanisms. Proteins are among the main targets for oxidants due to their high rate constants for several reactions with ROS and their abundance in biological systems. Protein damage has an important influence on cellular viability since most protein damage is non-repairable, and has deleterious consequences on protein structure and function. In addition, damaged and modified proteins can form cross-links and provide a basis for many senescence-associated alterations and may contribute to a range of human pathologies. Two proteolytic systems are responsible to ensure the maintenance of cellular functions: the proteasomal (UPS) and the lysosomal system. Those degrading systems provide a last line of antioxidative protection, removing irreversible damaged proteins and recycling amino acids for the continuous protein synthesis. But during aging, both systems are affected and their proteolytic activity declines significantly. Here we highlight the recent advantages in the understanding of protein oxidation and the fate of these damaged proteins during aging. This article is part of a Special Issue entitled: Posttranslational Protein modifications in biology and Medicine.
Collapse
Affiliation(s)
- Annika Höhn
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich Schiller University Jena, 07743 Jena, Germany
| | | | | |
Collapse
|
24
|
Joglekar MM, Panaskar SN, Chougale AD, Kulkarni MJ, Arvindekar AU. A novel mechanism for antiglycative action of limonene through stabilization of protein conformation. MOLECULAR BIOSYSTEMS 2013; 9:2463-72. [DOI: 10.1039/c3mb00020f] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Nonenzymatic glycosylation: A biochemical link between chronic hyperglycemia and pathophysiologic processes associated with diabetic complications and aging related debilities. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.biomag.2012.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
26
|
Yellowlees Douglas J, Bhatwadekar AD, Li Calzi S, Shaw LC, Carnegie D, Caballero S, Li Q, Stitt AW, Raizada MK, Grant MB. Bone marrow-CNS connections: implications in the pathogenesis of diabetic retinopathy. Prog Retin Eye Res 2012; 31:481-94. [PMID: 22609081 DOI: 10.1016/j.preteyeres.2012.04.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/25/2012] [Accepted: 04/26/2012] [Indexed: 12/13/2022]
Abstract
Diabetic retinopathy is the fourth most common cause of blindness in adults. Current therapies, including anti-VEGF therapy, have partial efficacy in arresting the progression of proliferative diabetic retinopathy and diabetic macular edema. This review provides an overview of a novel, innovative approach to viewing diabetic retinopathy as the result of an inflammatory cycle that affects the bone marrow (BM) and the central and sympathetic nervous systems. Diabetes associated inflammation may be the result of BM neuropathy which skews haematopoiesis towards generation of increased inflammatory cells but also reduces production of endothelial progenitor cells responsible for maintaining healthy endothelial function and renewal. The resulting systemic inflammation further impacts the hypothalamus, promoting insulin resistance and diabetes, and initiates an inflammatory cascade that adversely impacts both macrovascular and microvascular complications, including diabetic retinopathy (DR). This review examines the idea of using anti-inflammatory agents that cross not only the blood-retinal barrier to enter the retina but also have the capability to target the central nervous system and cross the blood-brain barrier to reduce neuroinflammation. This neuroinflammation in key sympathetic centers serves to not only perpetuate BM pathology but promote insulin resistance which is characteristic of type 2 diabetic patients (T2D) but is also seen in T1D. A case series of morbidly obese T2D patients with retinopathy and neuropathy treated with minocycline, a well-tolerated antibiotic that crosses both the blood-retina and blood-brain barrier is presented. Our results indicates that minocycine shows promise for improving visual acuity, reducing pain from peripheral neuropathy, promoting weight loss and improving blood pressure control and we postulate that these observed beneficial effects are due to a reduction of chronic inflammation.
Collapse
Affiliation(s)
- Jane Yellowlees Douglas
- Clinical and Translational Science Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Leong NL, Hurng JM, Djomehri SI, Gansky SA, Ryder MI, Ho SP. Age-related adaptation of bone-PDL-tooth complex: Rattus-Norvegicus as a model system. PLoS One 2012; 7:e35980. [PMID: 22558292 PMCID: PMC3340399 DOI: 10.1371/journal.pone.0035980] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/26/2012] [Indexed: 01/18/2023] Open
Abstract
Functional loads on an organ induce tissue adaptations by converting mechanical energy into chemical energy at a cell-level. The transducing capacity of cells alters physico-chemical properties of tissues, developing a positive feedback commonly recognized as the form-function relationship. In this study, organ and tissue adaptations were mapped in the bone-tooth complex by identifying and correlating biomolecular expressions to physico-chemical properties in rats from 1.5 to 15 months. However, future research using hard and soft chow over relevant age groups would decouple the function related effects from aging affects. Progressive curvature in the distal root with increased root resorption was observed using micro X-ray computed tomography. Resorption was correlated to the increased activity of multinucleated osteoclasts on the distal side of the molars until 6 months using tartrate resistant acid phosphatase (TRAP). Interestingly, mononucleated TRAP positive cells within PDL vasculature were observed in older rats. Higher levels of glycosaminoglycans were identified at PDL-bone and PDL-cementum entheses using alcian blue stain. Decreasing biochemical gradients from coronal to apical zones, specifically biomolecules that can induce osteogenic (biglycan) and fibrogenic (fibromodulin, decorin) phenotypes, and PDL-specific negative regulator of mineralization (asporin) were observed using immunohistochemistry. Heterogeneous distribution of Ca and P in alveolar bone, and relatively lower contents at the entheses, were observed using energy dispersive X-ray analysis. No correlation between age and microhardness of alveolar bone (0.7 ± 0.1 to 0.9 ± 0.2 GPa) and cementum (0.6 ± 0.1 to 0.8 ± 0.3 GPa) was observed using a microindenter. However, hardness of cementum and alveolar bone at any given age were significantly different (P<0.05). These observations should be taken into account as baseline parameters, during development (1.5 to 4 months), growth (4 to 10 months), followed by a senescent phase (10 to 15 months), from which deviations due to experimentally induced perturbations can be effectively investigated.
Collapse
Affiliation(s)
- Narita L. Leong
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| | - Jonathan M. Hurng
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| | - Sabra I. Djomehri
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| | - Stuart A. Gansky
- Division of Oral Epidemiology & Dental Public Health, Department of Preventive and Restorative Dental Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Mark I. Ryder
- Division of Periodontology, Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Sunita P. Ho
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
28
|
Hegab Z, Gibbons S, Neyses L, Mamas MA. Role of advanced glycation end products in cardiovascular disease. World J Cardiol 2012; 4:90-102. [PMID: 22558488 PMCID: PMC3342583 DOI: 10.4330/wjc.v4.i4.90] [Citation(s) in RCA: 238] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 04/04/2012] [Accepted: 04/10/2012] [Indexed: 02/06/2023] Open
Abstract
Advanced glycation end products (AGEs) are produced through the non enzymatic glycation and oxidation of proteins, lipids and nucleic acids. Enhanced formation of AGEs occurs particularly in conditions associated with hyperglycaemia such as diabetes mellitus (DM). AGEs are believed to have a key role in the development and progression of cardiovascular disease in patients with DM through the modification of the structure, function and mechanical properties of tissues through crosslinking intracellular as well as extracellular matrix proteins and through modulating cellular processes through binding to cell surface receptors [receptor for AGEs (RAGE)]. A number of studies have shown a correlation between serum AGE levels and the development and severity of heart failure (HF). Moreover, some studies have suggested that therapies targeted against AGEs may have therapeutic potential in patients with HF. The purpose of this review is to discuss the role of AGEs in cardiovascular disease and in particular in heart failure, focussing on both cellular mechanisms of action as well as highlighting how targeting AGEs may represent a novel therapeutic strategy in the treatment of HF.
Collapse
Affiliation(s)
- Zeinab Hegab
- Zeinab Hegab, Stephen Gibbons, Ludwig Neyses, Mamas A Mamas, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9WL, United Kingdom
| | | | | | | |
Collapse
|
29
|
Blakytny R, Spraul M, Jude EB. Review: The diabetic bone: a cellular and molecular perspective. INT J LOW EXTR WOUND 2011; 10:16-32. [PMID: 21444607 DOI: 10.1177/1534734611400256] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
With the increasing worldwide prevalence of diabetes the resulting complications, their consequences and treatment will lead to a greater social and financial burden on society. One of the many organs to be affected is bone. Loss of bone is observed in type 1 diabetes, in extreme cases mirroring osteoporosis, thus a greater risk of fracture. In the case of type 2 diabetes, both a loss and an increase of bone has been observed, although in both cases the quality of the bone overall was poorer, again leading to a greater risk of fracture. Once a fracture has occurred, healing is delayed in diabetes, including nonunion. The reasons leading to such changes in the state of the bone and fracture healing in diabetes is under investigation, including at the cellular and the molecular levels. In comparison with our knowledge of events in normal bone homeostasis and fracture healing, that for diabetes is much more limited, particularly in patients. However, progress is being made, especially with the use of animal models for both diabetes types. Identifying the molecular and cellular changes in the bone in diabetes and understanding how they arise will allow for targeted intervention to improve diabetic bone, thus helping to counter conditions such as Charcot foot as well as preventing fracture and accelerating healing when a fracture does occur.
Collapse
|
30
|
El-Bikai R, Welman M, Margaron Y, Côté JF, Macqueen L, Buschmann MD, Fahmi H, Shi Q, Maghni K, Fernandes JC, Benderdour M. Perturbation of adhesion molecule-mediated chondrocyte-matrix interactions by 4-hydroxynonenal binding: implication in osteoarthritis pathogenesis. Arthritis Res Ther 2010; 12:R201. [PMID: 20977750 PMCID: PMC2991038 DOI: 10.1186/ar3173] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 08/13/2010] [Accepted: 10/26/2010] [Indexed: 12/12/2022] Open
Abstract
Introduction Objectives were to investigate whether interactions between human osteoarthritic chondrocytes and 4-hydroxynonenal (HNE)-modified type II collagen (Col II) affect cell phenotype and functions and to determine the protective role of carnosine (CAR) treatment in preventing these effects. Methods Human Col II was treated with HNE at different molar ratios (MR) (1:20 to 1:200; Col II:HNE). Articular chondrocytes were seeded in HNE/Col II adduct-coated plates and incubated for 48 hours. Cell morphology was studied by phase-contrast and confocal microscopy. Adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and α1β1 integrin at protein and mRNA levels were quantified by Western blotting, flow cytometry and real-time reverse transcription-polymerase chain reaction. Cell death, caspases activity, prostaglandin E2 (PGE2), metalloproteinase-13 (MMP-13), mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB) were assessed by commercial kits. Col II, cyclooxygenase-2 (COX-2), MAPK, NF-κB-p65 levels were analyzed by Western blotting. The formation of α1β1 integrin-focal adhesion kinase (FAK) complex was revealed by immunoprecipitation. Results Col II modification by HNE at MR approximately 1:20, strongly induced ICAM-1, α1β1 integrin and MMP-13 expression as well as extracellular signal-regulated kinases 1 and 2 (ERK1/2) and NF-κB-p65 phosphorylation without impacting cell adhesion and viability or Col II expression. However, Col II modification with HNE at MR approximately 1:200, altered chondrocyte adhesion by evoking cell death and caspase-3 activity. It inhibited α1β1 integrin and Col II expression as well as ERK1/2 and NF-κB-p65 phosphorylation, but, in contrast, markedly elicited PGE2 release, COX-2 expression and p38 MAPK phosphorylation. Immunoprecipitation assay revealed the involvement of FAK in cell-matrix interactions through the formation of α1β1 integrin-FAK complex. Moreover, the modification of Col II by HNE at a 1:20 or approximately 1:200 MR affects parameters of the cell shape. All these effects were prevented by CAR, an HNE-trapping drug. Conclusions Our novel findings indicate that HNE-binding to Col II results in multiple abnormalities of chondrocyte phenotype and function, suggesting its contribution in osteoarthritis development. CAR was shown to be an efficient HNE-snaring agent capable of counteracting these outcomes.
Collapse
Affiliation(s)
- Rana El-Bikai
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Department of Surgery, University of Montreal, 5400 Gouin Blvd, West, Montreal, QC H4J 1C5, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zmatliková Z, Sedláková P, Lacinová K, Eckhardt A, Pataridis S, Mikšík I. Non-enzymatic posttranslational modifications of bovine serum albumin by oxo-compounds investigated by high-performance liquid chromatography-mass spectrometry and capillary zone electrophoresis-mass spectrometry. J Chromatogr A 2010; 1217:8009-15. [PMID: 20828700 DOI: 10.1016/j.chroma.2010.08.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 08/04/2010] [Accepted: 08/06/2010] [Indexed: 02/05/2023]
Abstract
Non-enzymatic posttranslational modifications of bovine serum albumin (BSA) by various oxo-compounds (glucose, ribose, glyoxal and glutardialdehyde) have been investigated using high-performance liquid chromatography (HPLC) and capillary zone electrophoresis (CZE). Both of these methods used mass spectrometric (MS) detection. Three enzymes (trypsin, pepsin, proteinase K) were used to digest glycated BSA. The extent of modification depended on the selected oxo-compound. Reactivity increased progressively from glucose to glutardialdehyde (glucose<ribose<glyoxal<glutardialdehyde). Carboxymethylation of lysine (CML) was the main type of modification detected. The HPLC/MS method achieved higher coverage and a larger amount of CML was identified compared to CZE/MS.
Collapse
Affiliation(s)
- Zdeňka Zmatliková
- Institute of Physiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | | | | | | | | | | |
Collapse
|
32
|
Yuen A, Laschinger C, Talior I, Lee W, Chan M, Birek J, Young EW, Sivagurunathan K, Won E, Simmons CA, McCulloch C. Methylglyoxal-modified collagen promotes myofibroblast differentiation. Matrix Biol 2010; 29:537-48. [DOI: 10.1016/j.matbio.2010.04.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 04/13/2010] [Accepted: 04/15/2010] [Indexed: 01/09/2023]
|
33
|
Aldose reductase deficiency improves Wallerian degeneration and nerve regeneration in diabetic thy1-YFP mice. J Neuropathol Exp Neurol 2010; 69:294-305. [PMID: 20142761 DOI: 10.1097/nen.0b013e3181d26487] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
This study examined the role of aldose reductase (AR) in diabetes-associated impaired nerve regeneration using thy1-YFP (YFP) mice. Sciatic nerves of nondiabetic and streptozotocin-induced diabetic AR(+/+)YFP and AR(-/-)YFP mice were transected after 4 weeks of diabetes. Wallerian degeneration and nerve regeneration were evaluated at 1 and 2 weeks postaxotomy by fluorescence microscopy. Motor nerve conduction velocity recovery and regenerating nerve morphometric parameters were determined at 10 and 20 weeks, respectively. There was no difference in the extent of Wallerian degeneration, size of regenerating stump, motor nerve conduction velocity recovery, or caliber of regenerating fibers between nondiabetic AR(+/+)YFP and AR(-/-)YFP mice. In diabetic AR(+/+)YFP mice, Wallerian degeneration was delayed, associated with slower macrophage invasion and abnormal vascularization. Those mice had smaller regenerating stumps, slower motor nerve conduction velocity, and smaller regenerating fibers compared with nondiabetic mice. These features of impaired nerve regeneration were largely attenuated in diabetic AR(-/-)YFP mice. Retarded macrophage invasion and vascularization associated with Wallerian degeneration were normalized in diabetic AR(-/-)YFP mice. These results indicate that AR plays an important role in diabetes-associated impaired nerve regeneration, in part by affecting vascularization and macrophage invasion during Wallerian degeneration. The thy1-YFP mice are valuable tools for further investigation of the mechanism of diabetes-associated nerve regeneration.
Collapse
|
34
|
Viguet-Carrin S, Follet H, Gineyts E, Roux JP, Munoz F, Chapurlat R, Delmas PD, Bouxsein ML. Association between collagen cross-links and trabecular microarchitecture properties of human vertebral bone. Bone 2010; 46:342-7. [PMID: 19836004 DOI: 10.1016/j.bone.2009.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 09/29/2009] [Accepted: 10/01/2009] [Indexed: 01/22/2023]
Abstract
UNLABELLED It has been suggested that age-related deterioration in trabecular microarchitecture and changes in collagen cross-link concentrations may contribute to skeletal fragility. To further explore this hypothesis, we determined the relationships among trabecular bone volume fraction (BV/TV), microarchitecture, collagen cross-link content, and bone turnover in human vertebral trabecular bone. Trabecular bone specimens from L2 vertebrae were collected from 51 recently deceased donors (54-95 years of age; 20 men and 30 women). Trabecular bone volume and microarchitecture was assessed by microCT and bone formation, reflected by osteoid surface (OS/BS, %), was measured by 2D histomorphometry. Pyridinoline (PYD), deoxypyridinoline (DPD), pentosidine (PEN) and collagen content in the cancellous bone were analysed by high-performance liquid chromatography. Associations between variables were investigated by Pearson correlations and multiple regression models, which were constructed with BV/TV and collagen cross-links as explanatory variables and microarchitecture parameters as the dependent variables. RESULTS Microarchitecture parameters were modestly to strongly correlated with BV/TV (r(2)=0.10-0.71). The amount of mature enzymatic PYD and DPD cross-links were not associated with the microarchitecture, either before or after adjustment for BV/TV. However, there was a positive correlation between PEN content and trabecular number (r=0.45, p=0.001) and connectivity density (r=0.40, p=0.004), and a negative correlation between PEN content and trabecular separation (r=-0.29, p=0.04). In the multiple regression models including BV/TV, age and PEN content was still significantly associated with several of the microarchitecture variables. In summary, this study suggests a link between trabecular microarchitecture and the collagen cross-link profile. As PEN reflects non-enzymatic glycation of collagen and generally increases with bone age, the association between PEN and trabecular architecture suggests that the preserved trabeculae may contain mainly old bone and have undergone little remodeling. Thus, vertebral fragility may not only be due to alterations in bone architecture but also to modification of collagen cross-link patterns thereby influencing bone's mechanical behavior.
Collapse
Affiliation(s)
- S Viguet-Carrin
- INSERM Research Unit 831 and Claude Bernard University of Lyon, Lyon, France.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Negre-Salvayre A, Salvayre R, Augé N, Pamplona R, Portero-Otín M. Hyperglycemia and glycation in diabetic complications. Antioxid Redox Signal 2009; 11:3071-109. [PMID: 19489690 DOI: 10.1089/ars.2009.2484] [Citation(s) in RCA: 269] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is a multifactorial disease, classically influenced by genetic determinants of individual susceptibility and by environmental accelerating factors, such as lifestyle. It is considered a major health concern,as its incidence is increasing at an alarming rate, and the high invalidating effects of its long-term complications affect macro- and microvasculature, heart, kidney, eye, and nerves. Increasing evidence indicates that hyperglycemia is the initiating cause of the tissue damage occurring in diabetes, either through repeated acute changes in cellular glucose metabolism, or through the long-term accumulation of glycated biomolecules and advanced glycation end products (AGEs). AGEs represent a heterogeneous group of chemical products resulting from a nonenzymatic reaction between reducing sugars and proteins, lipids, nucleic acids, or a combination of these.The glycation process (glucose fixation) affects circulating proteins (serum albumin, lipoprotein, insulin, hemoglobin),whereas the formation of AGEs implicates reactive intermediates such as methylglyoxal. AGEs form cross-links on long-lived extracellular matrix proteins or react with their specific receptor RAGE, resulting inoxidative stress and proinflammatory signaling implicated in endothelium dysfunction, arterial stiffening, and microvascular complications. This review summarizes the mechanism of glycation and of AGEs formation and the role of hyperglycemia, AGEs, and oxidative stress in the pathophysiology of diabetic complications.
Collapse
|
36
|
Sassi-Gaha S, Loughlin DT, Kappler F, Schwartz ML, Su B, Tobia AM, Artlett CM. Two dicarbonyl compounds, 3-deoxyglucosone and methylglyoxal, differentially modulate dermal fibroblasts. Matrix Biol 2009; 29:127-34. [PMID: 19800404 DOI: 10.1016/j.matbio.2009.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 09/14/2009] [Accepted: 09/21/2009] [Indexed: 11/26/2022]
Abstract
Advanced glycation endproducts accumulate on long-lived proteins such as collagens as a function of diet and age and mediate the cross-linking of those proteins causing changes in collagen pathophysiology resulting in the disruption of normal collagen matrix remodeling. Two commonly studied advanced glycation endproduct precursors 3-deoxyglucosone and methylglyoxal were investigated for their role in the modification of collagen and on extracellular matrix expression. Fibroblasts cultured on methylglyoxal cross-linked matrices increased the expression of collagen, active TGF-beta1, beta1-integrin, and decreased Smad7; whereas 3-deoxyglucosone decreased collagen, active TGF-beta1, beta1-integrin but increased Smad7. Purified collagen modified by 3-deoxyglucosone or methylglyoxal had different molecular weights; methylglyoxal increased the apparent molecular weight by approximately 20 kDa, whereas 3-deoxyglucosone did not. The differences in collagen expression by 3-deoxyglucosone and methylglyoxal raise the provocative idea that a genetic or environmental background leading to the predominance of one of these advanced glycation endproduct precursors may precipitate a fibrotic or chronic wound in susceptible individuals, particularly in the diabetic.
Collapse
Affiliation(s)
- Sihem Sassi-Gaha
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Beltramo E, Nizheradze K, Berrone E, Tarallo S, Porta M. Thiamine and benfotiamine prevent apoptosis induced by high glucose-conditioned extracellular matrix in human retinal pericytes. Diabetes Metab Res Rev 2009; 25:647-56. [PMID: 19768736 DOI: 10.1002/dmrr.1008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Early and selective loss of pericytes and thickening of the basement membrane are hallmarks of diabetic retinopathy. We reported reduced adhesion, but no changes in apoptosis, of bovine retinal pericytes cultured on extracellular matrix (ECM) produced by endothelial cells in high glucose (HG). Since human and bovine pericytes may behave differently in conditions mimicking the diabetic milieu, we verified the behaviour of human retinal pericytes cultured on HG-conditioned ECM. METHODS Pericytes were cultured in physiological/HG on ECM produced by human umbilical vein endothelial cells in physiological/HG, alone or in the presence of thiamine and benfotiamine. Adhesion, proliferation, apoptosis, p53 and Bcl-2/Bax ratio (mRNA levels and protein concentrations) were measured in wild-type and immortalized human pericytes. RESULTS Both types of pericytes adhered less to HG-conditioned ECM and plastic than to physiological glucose-conditioned ECM. DNA synthesis was impaired in pericytes cultured in HG on the three different surfaces but there were no differences in proliferation. DNA fragmentation and Bcl-2/Bax ratio were greatly enhanced by HG-conditioned ECM in pericytes kept in both physiological and HG. Addition of thiamine and benfotiamine to HG during ECM production completely prevented these damaging effects. CONCLUSIONS Apoptosis is strongly increased in pericytes cultured on ECM produced by endothelium in HG, probably due to impairment of the Bcl-2/Bax ratio. Thiamine and benfotiamine completely revert this effect. This behaviour is therefore completely different from that of bovine pericytes, underlining the importance of establishing species-specific cell models to study the mechanisms of diabetic retinopathy.
Collapse
Affiliation(s)
- Elena Beltramo
- Laboratory of Diabetic Retinopathy, Department of Internal Medicine, University of Turin, Torino, Italy.
| | | | | | | | | |
Collapse
|
38
|
Peppa M, Stavroulakis P, Raptis SA. Advanced glycoxidation products and impaired diabetic wound healing. Wound Repair Regen 2009; 17:461-72. [PMID: 19614910 DOI: 10.1111/j.1524-475x.2009.00518.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Impaired wound healing is an important diabetic complication associated with increased morbidity and mortality. It appears to be the net result of micro- and macrovascular disease. Diabetic neuropathy and the resulting loss of protective sensation (LOPS) has been recognized as one of the major causes for delayed healing in diabetic foot ulcer patients. In addition, hyperglycemia and a number of hyperglycemia-related factors have been linked to impaired diabetic wound healing, including advanced glycation end products (AGE). A large body of evidence from in vitro and in vivo studies and also data from studies using anti-AGE agents, indicate that AGE may play a role in the pathogenesis of impaired diabetic wound healing. AGE affect the wound healing process either directly by their interference with a variety of components involved or indirectly through their association with diabetic neuropathy and/or angiopathy. However, further studies need to be performed, mostly clinical studies, to evaluate the exact role of AGE in the impaired diabetic wound healing, suggesting new therapeutic approaches.
Collapse
Affiliation(s)
- Melpomeni Peppa
- Endocrine Unit, Second Department of Internal Medicine-Propaedeutic, Research Institute and Diabetes Center, Athens University Medical School, Attikon University Hospital, Athens, Greece.
| | | | | |
Collapse
|
39
|
Murdaugh L, Dillon J, Gaillard E. Modifications to the basement membrane protein laminin using glycolaldehyde and A2E: A model for aging in Bruch's membrane. Exp Eye Res 2009; 89:187-92. [DOI: 10.1016/j.exer.2009.03.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 03/14/2009] [Accepted: 03/17/2009] [Indexed: 10/20/2022]
|
40
|
Davies CA, Herrick AL, Cordingley L, Freemont AJ, Jeziorska M. Expression of advanced glycation end products and their receptor in skin from patients with systemic sclerosis with and without calcinosis. Rheumatology (Oxford) 2009; 48:876-82. [DOI: 10.1093/rheumatology/kep151] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
41
|
Liao H, Zakhaleva J, Chen W. Cells and tissue interactions with glycated collagen and their relevance to delayed diabetic wound healing. Biomaterials 2009; 30:1689-96. [PMID: 19157537 DOI: 10.1016/j.biomaterials.2008.11.038] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 11/18/2008] [Indexed: 11/17/2022]
Abstract
Dermal accumulation of advanced glycation end products (AGEs) has increasingly been implicated as the underlying cause of delayed diabetic wound healing. Devising an in vitro model to adequately mimic glycated tissues will facilitate investigation into the mechanism of glycation in conjunction with exploration of new approaches or improvement of current therapies for treating diabetic chronic wounds. Collagen matrices were artificially glycated and the presence of AGEs was demonstrated by immunostaining. Both the mechanical properties of the collagen matrices and their interactions with fibroblasts (morphology, attachment, proliferation, and migration) were altered after glycation, moreover, there was evidence of impairment on extracellular matrix (ECM) remodeling as well as inhibition of cell-induced material contraction. The actin cytoskeletons of the fibroblasts residing in the glycated collagen matrices were reorganized. In vivo mice full-thickness dermal wound models implanted with glycated collagen matrices showed delayed wound healing response. Thus, the glycated collagen matrix is an adequate in vitro model to mimic glycated tissues and could serve as a facile experimental tool to investigate the mechanism of glycation in conjunction with exploration of new approaches or improvement of current therapies for treating diabetic wounds.
Collapse
Affiliation(s)
- Huijuan Liao
- Department of Biomedical Engineering, State University of New York - Stony Brook, Health Science Center, 11794-8181, USA
| | | | | |
Collapse
|
42
|
Reigle KL, Di Lullo G, Turner KR, Last JA, Chervoneva I, Birk DE, Funderburgh JL, Elrod E, Germann MW, Surber C, Sanderson RD, San Antonio JD. Non-enzymatic glycation of type I collagen diminishes collagen-proteoglycan binding and weakens cell adhesion. J Cell Biochem 2008; 104:1684-98. [PMID: 18348167 DOI: 10.1002/jcb.21735] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Non-enzymatic glycation of type I collagen occurs in aging and diabetes, and may affect collagen solubility, charge, polymerization, and intermolecular interactions. Proteoglycans(1) (PGs) bind type I collagen and are proposed to regulate fibril assembly, function, and cell-collagen interactions. Moreover, on the collagen fibril a keratan sulfate (KS) PG binding region overlaps with preferred collagen glycation sites. Thus, we examined the effect of collagen modified by simple glycation on PG-collagen interactions. By affinity coelectrophoresis (ACE), we found reduced affinities of heparin and KSPGs for glycated but not normal collagen, whereas the dermatan sulfate (DS)PGs decorin and biglycan bound similarly to both, and that the affinity of heparin for normal collagen decreased with increasing pH. Circular dichroism (CD) spectroscopy revealed normal and glycated collagens to assume triple helical conformations, but heparin addition caused precipitation and decreased triple helical content-effects that were more marked with glycated collagen. A spectrophotometric assay revealed slower polymerization of glycated collagen. However, ultrastructural analyses indicated that fibrils assembled from normal and glycated collagen exhibited normal periodicity, and had similar structures and comparable diameter distributions. B-cells expressing the cell surface heparan sulfate PG syndecan-1 adhered well to normal but not glycated collagen, and endothelial cell migration was delayed on glycated collagen. We speculate that glycation diminishes the electrostatic interactions between type I collagen and PGs, and may interfere with core protein-collagen associations for KSPGs but not DSPGs. Therefore in vivo, collagen glycation may weaken PG-collagen interactions, thereby disrupting matrix integrity and cell-collagen interactions, adhesion, and migration.
Collapse
Affiliation(s)
- Kristin L Reigle
- Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania 19107-5099, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sweeney SM, Orgel JP, Fertala A, McAuliffe JD, Turner KR, Di Lullo GA, Chen S, Antipova O, Perumal S, Ala-Kokko L, Forlino A, Cabral WA, Barnes AM, Marini JC, Antonio JDS. Candidate cell and matrix interaction domains on the collagen fibril, the predominant protein of vertebrates. J Biol Chem 2008; 283:21187-97. [PMID: 18487200 PMCID: PMC2475701 DOI: 10.1074/jbc.m709319200] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 04/11/2008] [Indexed: 11/06/2022] Open
Abstract
Type I collagen, the predominant protein of vertebrates, polymerizes with type III and V collagens and non-collagenous molecules into large cable-like fibrils, yet how the fibril interacts with cells and other binding partners remains poorly understood. To help reveal insights into the collagen structure-function relationship, a data base was assembled including hundreds of type I collagen ligand binding sites and mutations on a two-dimensional model of the fibril. Visual examination of the distribution of functional sites, and statistical analysis of mutation distributions on the fibril suggest it is organized into two domains. The "cell interaction domain" is proposed to regulate dynamic aspects of collagen biology, including integrin-mediated cell interactions and fibril remodeling. The "matrix interaction domain" may assume a structural role, mediating collagen cross-linking, proteoglycan interactions, and tissue mineralization. Molecular modeling was used to superimpose the positions of functional sites and mutations from the two-dimensional fibril map onto a three-dimensional x-ray diffraction structure of the collagen microfibril in situ, indicating the existence of domains in the native fibril. Sequence searches revealed that major fibril domain elements are conserved in type I collagens through evolution and in the type II/XI collagen fibril predominant in cartilage. Moreover, the fibril domain model provides potential insights into the genotype-phenotype relationship for several classes of human connective tissue diseases, mechanisms of integrin clustering by fibrils, the polarity of fibril assembly, heterotypic fibril function, and connective tissue pathology in diabetes and aging.
Collapse
Affiliation(s)
- Shawn M. Sweeney
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Joseph P. Orgel
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Andrzej Fertala
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Jon D. McAuliffe
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Kevin R. Turner
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Gloria A. Di Lullo
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Steven Chen
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Olga Antipova
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Shiamalee Perumal
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Leena Ala-Kokko
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Antonella Forlino
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Wayne A. Cabral
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Aileen M. Barnes
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - Joan C. Marini
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| | - James D. San Antonio
- Cardiovascular Institute, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, the
Center for Synchrotron Radiation Research
and Instrumentation, Department of Biological, Chemical, and Physical
Sciences, Illinois Institute of Technology, Chicago, Illinois 60616, the
Department of Dermatology and Cutaneous
Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, the
Department of Statistics, Wharton School,
University of Pennsylvania, Philadelphia 19104, Pennsylvania, the
Chicago Medical School, North Chicago,
Illinois 60064, the Collagen Research
Unit, Biocenter and Department of Medical Biochemistry and Molecular Biology,
University of Oulu, Oulu, Finland,
Connective Tissue Gene Tests, Allentown,
Pennsylvania 18103, the Department of
Biochemistry A. Castellani, University of Pavia, Pavia, Italy, the
Bone and Extracellular Matrix Branch,
Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland 20892, and the
Cardeza Foundation for Hematologic
Research and Department of Medicine, Thomas Jefferson University,
Philadelphia, Pennsylvania 19107
| |
Collapse
|
44
|
Abstract
Neurons have a constantly high glucose demand, and unlike muscle cells they cannot accommodate episodic glucose uptake under the influence of insulin. Neuronal glucose uptake depends on the extracellular concentration of glucose, and cellular damage can ensue after persistent episodes of hyperglycaemia--a phenomenon referred to as glucose neurotoxicity. This article reviews the pathophysiological manifestation of raised glucose in neurons and how this can explain the major components of diabetic neuropathy.
Collapse
Affiliation(s)
- David R Tomlinson
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| | | |
Collapse
|
45
|
Abstract
The function of the kidney, as well as its morphology, changes markedly with age. The glomerular filtration rate falls progressively, independent of overt pathology. Glomerular, vascular and accompanying parenchymal changes occur and other disorders associated with ageing, such as diabetes and hypertension, have a stochastic deleterious effect on both form and function. Declining renal function with age has important implications, not only for individual homeostasis but also for the use of drug therapy and for the receipt and donation of organs for transplantation. Molecular mechanisms and cellular changes underlying some of the functional and structural changes associated with ageing are becoming clearer, as are some of the ways in which genetic background, age and disease can combine to produce functional damage.
Collapse
Affiliation(s)
- J E Martin
- Pathology Group, Institute of Cell and Molecular Sciences, St Bartholomew's and the London Hospital School of Medicine and Dentistry, London, UK.
| | | |
Collapse
|
46
|
Abstract
In general terms, the recognized alterations in circulating humoral factors (hormones, cytokines, growth factors) that occur in ageing, coupled with innate cellular senescence exaggerated by the slow turnover of many connective tissue cell populations and the age-associated alterations in matrix molecule cross-linking, predispose the elderly to altered connective tissue biology. These changes can be profound, leading to poor mobility, altered ability to withstand cold, weakness and an increased risk of falls, fractures and age-associated 'degenerative' diseases, such as osteoarthritis and osteoporosis. As understanding of the causes of altered connective tissue function with age increases, it is becoming clearer that many of the predisposing factors (growth hormone, cytokines, load/life style) are potential targets for improving quality of life in the elderly.
Collapse
Affiliation(s)
- A J Freemont
- Division of Regenerative Medicine, The Medical School, The University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| | | |
Collapse
|
47
|
Tchaikovski V, Waltenberger J. Angiogenesis and Arteriogenesis in Diabetes Mellitus: Signal Transduction Defects as the Molecular Basis of Vascular Cell Dysfunction. THERAPEUTIC NEOVASCULARIZATION–QUO VADIS? 2007:33-73. [DOI: 10.1007/1-4020-5955-8_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
48
|
Chong SAC, Lee W, Arora PD, Laschinger C, Young EWK, Simmons CA, Manolson M, Sodek J, McCulloch CA. Methylglyoxal inhibits the binding step of collagen phagocytosis. J Biol Chem 2007; 282:8510-20. [PMID: 17229729 DOI: 10.1074/jbc.m609859200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Bacterial infection-induced fibrosis affects a wide variety of tissues, including the periodontium, but the mechanisms that dysregulate matrix turnover and mediate fibrosis are not defined. Since collagen turnover by phagocytosis is an important pathway for matrix remodeling, we studied the effect of the bacterial and eukaryotic cell metabolite, methylglyoxal (MGO), on the binding step of phagocytosis by periodontal fibroblasts. Type 1 collagen was treated with various concentrations of methylglyoxal, an important glucose metabolite that modifies Arg and Lys residues. The extent of MGO-induced modifications was authenticated by amino acid analysis, solubility, and cross-linking. Cells were incubated with fluorescent beads coated with collagen, and the percentage of phagocytic cells was estimated by flow cytometry. MGO inhibited collagen binding (20% of control for 10 mm MGO) in a time- and concentration-dependent manner. MGO-induced inhibition of binding was prevented by aminoguanidine, which blocks the formation of collagen cross-links. MGO reduced collagen binding strength and blocked intracellular calcium signaling. MGO modified the Arg residue in the critical alpha2beta1 integrin-binding recognition sequence of triple helical collagen peptides, whereas MGO-induced cross-linking of Lys residues played only a small role in binding inhibition. Thus, MGO modifications of Arg residues in collagen could be a key factor in the impaired degradation of collagen that promotes fibrosis in chronic infections, such as periodontitis.
Collapse
Affiliation(s)
- Sandra A C Chong
- Canadian Institutes of Health Research Group in Matrix Dynamics, University of Toronto, Toronto, Ontario M5S 3E2, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Cellular and extracellular proteins suffer significant damage in vivo by glycation. Physiological proteolysis of proteins damaged by glycation forms glycation free adducts that are released into plasma for urinary excretion. Inefficient elimination of these free adducts in uremia leads to their accumulation. In mild renal insufficiency, plasma glycation free adducts accumulated as renal clearance declined. In patients with end-stage renal disease, plasma glycation free adducts were increased up to 18-fold on peritoneal dialysis and up to 40-fold on hemodialysis. Glycation free adduct concentrations in peritoneal dialysate increased with dialysate dwell time, achieving concentrations in the dialysate higher than in plasma--suggesting that glycation adduct formation may occur in the peritoneal cavity and active transport into the peritoneal cavity may occur. In hemodialysis, plasma glycation free adducts equilibrated rapidly across the dialysis membrane, with both plasma and dialysate concentrations decreasing during a dialysis session. Therefore, protein glycation free adducts normally excreted efficiently in urine show profound mishandling and accumulation in chronic renal failure. Their accumulation may impair vascular cell function and contribute to morbidity and mortality in renal disease.
Collapse
Affiliation(s)
- Paul J Thornalley
- Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex, UK.
| |
Collapse
|
50
|
Avery NC, Bailey AJ. The effects of the Maillard reaction on the physical properties and cell interactions of collagen. ACTA ACUST UNITED AC 2006; 54:387-95. [PMID: 16962252 DOI: 10.1016/j.patbio.2006.07.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Accepted: 07/04/2006] [Indexed: 12/29/2022]
Abstract
The non-enzymic glycation of collagen occurs as its turnover decreases during maturation, with complex carbohydrates accumulating slowly and the end-products of these reactions being permanent. The nature of these advanced glycation end-reaction products (AGEs) can be categorised as: 1) cross-linking: intermolecular cross-linking may occur between two adjacent molecules and involve lysine to lysine or lysine to arginine residues. Several compounds have been characterised. They are believed to be located between the triple helical domains of adjacent molecules in the fibre resulting in major changes of the physical properties, primarily, fibre stiffness, thermal denaturation temperature and enzyme resistance, all of which increase slowly with age but the rate is accelerated in diabetes mellitus due to high glucose levels: 2) side-chain modifications: these changes alter the charge profile of the molecule affecting the interactions within the fibre and if they occur at specific sites can affect the cell-collagen interaction. Modification of arginine within the sites RGD and GFOGER recognised by the two specific integrins (alpha1beta2 and alpha2beta1) for collagen reduce cell interactions during turnover and for platelet interactions (alpha1beta2). These changes can ultimately affect repair of, for example, vascular damage and dermal wound healing in diabetes mellitus. Both types of modification are deleterious to the optimal properties of collagen as a supporting framework structure and as a controlling factor in cell matrix interactions. Glycation during ageing and diabetes is therefore responsible for malfunctioning of the diverse collagenous tissues throughout the body.
Collapse
Affiliation(s)
- N C Avery
- Collagen Research Group, University of Bristol, Langford, BS40 5DU Bristol, UK
| | | |
Collapse
|