1
|
Jarosławska J, Kordas B, Miłowski T, Juranek JK. Mammalian Diaphanous1 signalling in neurovascular complications of diabetes. Eur J Neurosci 2024; 59:2628-2645. [PMID: 38491850 DOI: 10.1111/ejn.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/18/2024] [Indexed: 03/18/2024]
Abstract
Over the past few decades, diabetes gradually has become one of the top non-communicable disorders, affecting 476.0 million in 2017 and is predicted to reach 570.9 million people in 2025. It is estimated that 70 to 100% of all diabetic patients will develop some if not all, diabetic complications over the course of the disease. Despite different symptoms, mechanisms underlying the development of diabetic complications are similar, likely stemming from deficits in both neuronal and vascular components supplying hyperglycaemia-susceptible tissues and organs. Diaph1, protein diaphanous homolog 1, although mainly known for its regulatory role in structural modification of actin and related cytoskeleton proteins, in recent years attracted research attention as a cytoplasmic partner of the receptor of advanced glycation end-products (RAGE) a signal transduction receptor, whose activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines in diabetes and its related complications. Both Diaph1 and RAGE are also a part of the RhoA signalling cascade, playing a significant role in the development of neurovascular disturbances underlying diabetes-related complications. In this review, based on the existing knowledge as well as compelling findings from our past and present studies, we address the role of Diaph1 signalling in metabolic stress and neurovascular degeneration in diabetic complications. In light of the most recent developments in biochemical, genomic and transcriptomic research, we describe current theories on the aetiology of diabetes complications, highlighting the function of the Diaph1 signalling system and its role in diabetes pathophysiology.
Collapse
Affiliation(s)
- Julia Jarosławska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Tadeusz Miłowski
- Department of Emergency Medicine, School of Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Judyta K Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
2
|
Sourris KC, Ding Y, Maxwell SS, Al-Sharea A, Kantharidis P, Mohan M, Rosado CJ, Penfold SA, Haase C, Xu Y, Forbes JM, Crawford S, Ramm G, Harcourt BE, Jandeleit-Dahm K, Advani A, Murphy AJ, Timmermann DB, Karihaloo A, Knudsen LB, El-Osta A, Drucker DJ, Cooper ME, Coughlan MT. Glucagon-like peptide-1 receptor signaling modifies the extent of diabetic kidney disease through dampening the receptor for advanced glycation end products-induced inflammation. Kidney Int 2024; 105:132-149. [PMID: 38069998 DOI: 10.1016/j.kint.2023.09.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 01/07/2024]
Abstract
Glucagon like peptide-1 (GLP-1) is a hormone produced and released by cells of the gastrointestinal tract following meal ingestion. GLP-1 receptor agonists (GLP-1RA) exhibit kidney-protective actions through poorly understood mechanisms. Here we interrogated whether the receptor for advanced glycation end products (RAGE) plays a role in mediating the actions of GLP-1 on inflammation and diabetic kidney disease. Mice with deletion of the GLP-1 receptor displayed an abnormal kidney phenotype that was accelerated by diabetes and improved with co-deletion of RAGE in vivo. Activation of the GLP-1 receptor pathway with liraglutide, an anti-diabetic treatment, downregulated kidney RAGE, reduced the expansion of bone marrow myeloid progenitors, promoted M2-like macrophage polarization and lessened markers of kidney damage in diabetic mice. Single cell transcriptomics revealed that liraglutide induced distinct transcriptional changes in kidney endothelial, proximal tubular, podocyte and macrophage cells, which were dominated by pathways involved in nutrient transport and utilization, redox sensing and the resolution of inflammation. The kidney-protective action of liraglutide was corroborated in a non-diabetic model of chronic kidney disease, the subtotal nephrectomised rat. Thus, our findings identify a novel glucose-independent kidney-protective action of GLP-1-based therapies in diabetic kidney disease and provide a valuable resource for exploring the cell-specific kidney transcriptional response ensuing from pharmacological GLP-1R agonism.
Collapse
Affiliation(s)
- Karly C Sourris
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia; Diabetes Complications Division, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia.
| | - Yi Ding
- Diabetes Complications Division, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia; Diabetes Complications Research, Novo Nordisk, Måløv, Denmark
| | - Scott S Maxwell
- Epigenetics in Human Health and Disease Program, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Annas Al-Sharea
- Haematopoiesis and Leukocyte Biology, Division of Immunometabolism, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Phillip Kantharidis
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia
| | - Muthukumar Mohan
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia
| | - Carlos J Rosado
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia
| | - Sally A Penfold
- Diabetes Complications Division, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Claus Haase
- Diabetes Complications Research, Novo Nordisk, Måløv, Denmark
| | - Yangsong Xu
- Haematopoiesis and Leukocyte Biology, Division of Immunometabolism, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Josephine M Forbes
- Mater Research Institute, the University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Simon Crawford
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Georg Ramm
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Brooke E Harcourt
- Murdoch Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Karin Jandeleit-Dahm
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michaels Hospital, Toronto, Ontario, Canada
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Division of Immunometabolism, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Anil Karihaloo
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington, USA
| | | | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mark E Cooper
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Monash University, Central Clinical School, Alfred Research Alliance, Melbourne, Victoria, Australia; Diabetes Complications Division, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Parkville, Victoria, Australia.
| |
Collapse
|
3
|
Li C, Yang W, Meng Y, Feng L, Sun L, Li Z, Liu X, Li M. Exploring the therapeutic mechanism of Banxia Xiexin Decoction in mild cognitive impairment and diabetes mellitus: a network pharmacology approach. Metab Brain Dis 2023; 38:2315-2325. [PMID: 37556042 DOI: 10.1007/s11011-023-01270-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/10/2023]
Abstract
The incidence of mild cognitive impairment (MCI) and diabetes mellitus (DM) is increasing year by year. Clinical findings show that Banxia Xiexin Decoction (BXD) can be combined to treat MCI and DM. However, the principle and mechanism of BXD in treating MCI and DM remain unclear. In this study, to explore the common mechanism of BXD in treating MCI and DM by using the method of network pharmacology. Traditional Chinese Medicine Systems Pharmacology Database (TCMSP) was used to screen the main active components of BXD, as well as to predict and screen its potential targets. Using Online Mendelian Inheritance in Man (OMIM), Therapeutic Target Database (TTD), DisGeNET, GeneCards to select the target proteins of two diseases, and intersecting the drug target and the disease target to obtain the common target of drug diseases, which is imported into cytoscape software to draw the network diagram of "drug components-target diseases" and the interaction network diagram between the common target proteins. According to the Database for Annotation, Visualization and Integrated Discovery (DAVID) database, we analyzed the common targets using two methods, gene ontology Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathway enrichment analysis and Gene Ontology (GO) function enrichment analysis, as well as studied the interaction mechanism of the two diseases, with the results validated using molecular docking. A total of 267 main active components of BXD were screened, together with the two diseases shared 233 common targets. The top five key targets identified by the topological analysis were TP53, AKT1, STAT3, TNF, and MAPK3. Go enrichment results indicated that it was primarily related to response to drug, extracellular space, enzyme binding, RNA polymerase II transcription factor activity, ligand-activated sequence-specific DNA binding. t KEGG enrichment pathway analysis identified 20 significant pathways, the majority of which are AGE-RAGE signaling pathways in diabetic complications, lipid and atherosclerosis, fluid shear stress and atherosclerosis, IL-17 signaling pathway, TNF signaling pathway, and so on. The results of molecular docking revealed that the key components of BXD, baicalein, licochalcone a, quercetin, and naringenin, had strong binding ability with core targets TP53, AKT1, STAT3, TNF, MAPK3. BXD can treat MCI and DM by multi-targets and multi-channels,and plays a role of "homotherapy for heteropathy" mainly through response to drug, positive regulation of gene expression, extracellular space and enzyme binding and other ways.
Collapse
Affiliation(s)
- Cong Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Wei Yang
- Neurology Department, Affiliated Hospital of the Changchun University of Chinese Medicine, Changchun, China
| | - Yubo Meng
- Neurology Department, Affiliated Hospital of the Changchun University of Chinese Medicine, Changchun, China
| | - Lina Feng
- Neurology Department, Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, China
| | - Linlin Sun
- Neurology Department, Affiliated Hospital of the Changchun University of Chinese Medicine, Changchun, China
| | - Zhenghong Li
- Research Department, Swiss University of Traditional Chinese Medicine, Bad Zurzach, Switzerland
| | - Xingfang Liu
- Research Department, Swiss University of Traditional Chinese Medicine, Bad Zurzach, Switzerland
| | - Mingquan Li
- Neurology Department, Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, China.
| |
Collapse
|
4
|
Reynaert NL, Vanfleteren LEGW, Perkins TN. The AGE-RAGE Axis and the Pathophysiology of Multimorbidity in COPD. J Clin Med 2023; 12:jcm12103366. [PMID: 37240472 DOI: 10.3390/jcm12103366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease of the airways and lungs due to an enhanced inflammatory response, commonly caused by cigarette smoking. Patients with COPD are often multimorbid, as they commonly suffer from multiple chronic (inflammatory) conditions. This intensifies the burden of individual diseases, negatively affects quality of life, and complicates disease management. COPD and comorbidities share genetic and lifestyle-related risk factors and pathobiological mechanisms, including chronic inflammation and oxidative stress. The receptor for advanced glycation end products (RAGE) is an important driver of chronic inflammation. Advanced glycation end products (AGEs) are RAGE ligands that accumulate due to aging, inflammation, oxidative stress, and carbohydrate metabolism. AGEs cause further inflammation and oxidative stress through RAGE, but also through RAGE-independent mechanisms. This review describes the complexity of RAGE signaling and the causes of AGE accumulation, followed by a comprehensive overview of alterations reported on AGEs and RAGE in COPD and in important co-morbidities. Furthermore, it describes the mechanisms by which AGEs and RAGE contribute to the pathophysiology of individual disease conditions and how they execute crosstalk between organ systems. A section on therapeutic strategies that target AGEs and RAGE and could alleviate patients from multimorbid conditions using single therapeutics concludes this review.
Collapse
Affiliation(s)
- Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Lowie E G W Vanfleteren
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Timothy N Perkins
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
5
|
Steckelings UM, Widdop RE, Sturrock ED, Lubbe L, Hussain T, Kaschina E, Unger T, Hallberg A, Carey RM, Sumners C. The Angiotensin AT 2 Receptor: From a Binding Site to a Novel Therapeutic Target. Pharmacol Rev 2022; 74:1051-1135. [PMID: 36180112 PMCID: PMC9553111 DOI: 10.1124/pharmrev.120.000281] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Discovered more than 30 years ago, the angiotensin AT2 receptor (AT2R) has evolved from a binding site with unknown function to a firmly established major effector within the protective arm of the renin-angiotensin system (RAS) and a target for new drugs in development. The AT2R represents an endogenous protective mechanism that can be manipulated in the majority of preclinical models to alleviate lung, renal, cardiovascular, metabolic, cutaneous, and neural diseases as well as cancer. This article is a comprehensive review summarizing our current knowledge of the AT2R, from its discovery to its position within the RAS and its overall functions. This is followed by an in-depth look at the characteristics of the AT2R, including its structure, intracellular signaling, homo- and heterodimerization, and expression. AT2R-selective ligands, from endogenous peptides to synthetic peptides and nonpeptide molecules that are used as research tools, are discussed. Finally, we summarize the known physiological roles of the AT2R and its abundant protective effects in multiple experimental disease models and expound on AT2R ligands that are undergoing development for clinical use. The present review highlights the controversial aspects and gaps in our knowledge of this receptor and illuminates future perspectives for AT2R research. SIGNIFICANCE STATEMENT: The angiotensin AT2 receptor (AT2R) is now regarded as a fully functional and important component of the renin-angiotensin system, with the potential of exerting protective actions in a variety of diseases. This review provides an in-depth view of the AT2R, which has progressed from being an enigma to becoming a therapeutic target.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert E Widdop
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Edward D Sturrock
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Lizelle Lubbe
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Tahir Hussain
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Elena Kaschina
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Thomas Unger
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Anders Hallberg
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert M Carey
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Colin Sumners
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| |
Collapse
|
6
|
Scavello F, Piacentini L, Castiglione S, Zeni F, Macrì F, Casaburo M, Vinci MC, Colombo GI, Raucci A. Effects of RAGE Deletion on the Cardiac Transcriptome during Aging. Int J Mol Sci 2022; 23:ijms231911130. [PMID: 36232442 PMCID: PMC9569842 DOI: 10.3390/ijms231911130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiac aging is characterized by increased cardiomyocyte hypertrophy, myocardial stiffness, and fibrosis, which enhance cardiovascular risk. The receptor for advanced glycation end-products (RAGE) is involved in several age-related diseases. RAGE knockout (Rage−/−) mice show an acceleration of cardiac dimension changes and interstitial fibrosis with aging. This study identifies the age-associated cardiac gene expression signature induced by RAGE deletion. We analyzed the left ventricle transcriptome of 2.5-(Young), 12-(Middle age, MA), and 21-(Old) months-old female Rage−/− and C57BL/6N (WT) mice. By comparing Young, MA, and Old Rage−/− versus age-matched WT mice, we identified 122, 192, and 12 differently expressed genes, respectively. Functional inference analysis showed that RAGE deletion is associated with: (i) down-regulation of genes involved in antigen processing and presentation of exogenous antigen, adaptive immune response, and cellular responses to interferon beta and gamma in Young animals; (ii) up-regulation of genes related to fatty acid oxidation, cardiac structure remodeling and cellular response to hypoxia in MA mice; (iii) up-regulation of few genes belonging to complement activation and triglyceride biosynthetic process in Old animals. Our findings show that the age-dependent cardiac phenotype of Rage−/− mice is associated with alterations of genes related to adaptive immunity and cardiac stress pathways.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Luca Piacentini
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Federica Macrì
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Manuel Casaburo
- Animal Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Maria Cristina Vinci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Gualtiero I. Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Correspondence: (G.I.C.); (A.R.); Tel.: +39-025-800-2464 (G.I.C.); +39-025-800-2802 (A.R.); Fax: +39-025-800-2342 (G.I.C. & A.R.)
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Animal Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Correspondence: (G.I.C.); (A.R.); Tel.: +39-025-800-2464 (G.I.C.); +39-025-800-2802 (A.R.); Fax: +39-025-800-2342 (G.I.C. & A.R.)
| |
Collapse
|
7
|
Advanced Glycation End Products (AGEs) and Chronic Kidney Disease: Does the Modern Diet AGE the Kidney? Nutrients 2022; 14:nu14132675. [PMID: 35807857 PMCID: PMC9268915 DOI: 10.3390/nu14132675] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/13/2022] Open
Abstract
Since the 1980s, chronic kidney disease (CKD) affecting all ages has increased by almost 25%. This increase may be partially attributable to lifestyle changes and increased global consumption of a “western” diet, which is typically energy dense, low in fruits and vegetables, and high in animal protein and ultra-processed foods. These modern food trends have led to an increase in the consumption of advanced glycation end products (AGEs) in conjunction with increased metabolic dysfunction, obesity and diabetes, which facilitates production of endogenous AGEs within the body. When in excess, AGEs can be pathological via both receptor-mediated and non-receptor-mediated pathways. The kidney, as a major site for AGE clearance, is particularly vulnerable to AGE-mediated damage and increases in circulating AGEs align with risk of CKD and all-cause mortality. Furthermore, individuals with significant loss of renal function show increased AGE burden, particularly with uraemia, and there is some evidence that AGE lowering via diet or pharmacological inhibition may be beneficial for CKD. This review discusses the pathways that drive AGE formation and regulation within the body. This includes AGE receptor interactions and pathways of AGE-mediated pathology with a focus on the contribution of diet on endogenous AGE production and dietary AGE consumption to these processes. We then analyse the contribution of AGEs to kidney disease, the evidence for dietary AGEs and endogenously produced AGEs in driving pathogenesis in diabetic and non-diabetic kidney disease and the potential for AGE targeted therapies in kidney disease.
Collapse
|
8
|
Sabbatinelli J, Castiglione S, Macrì F, Giuliani A, Ramini D, Vinci MC, Tortato E, Bonfigli AR, Olivieri F, Raucci A. Circulating levels of AGEs and soluble RAGE isoforms are associated with all-cause mortality and development of cardiovascular complications in type 2 diabetes: a retrospective cohort study. Cardiovasc Diabetol 2022; 21:95. [PMID: 35668468 PMCID: PMC9169316 DOI: 10.1186/s12933-022-01535-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Advanced glycation end-products (AGEs) and their interaction with the receptor for advanced glycation end-products (RAGE) play a pivotal role in the development and progression of type 2 diabetes. In this retrospective cohort study, we explored the association of circulating levels of soluble RAGE (sRAGE) isoforms, i.e., endogenous secretory esRAGE and cleaved cRAGE, AGEs and their respective ratios with 15-year all-cause mortality in type 2 diabetes. METHODS Baseline AGEs and sRAGE isoforms concentration were measured by ELISA in 362 patients with type 2 diabetes and in 125 age- and gender-matched healthy control subjects (CTR). Independent predictors of mortality were determined using Cox proportional-hazards models and used to build and validate a nomogram for all-cause mortality prediction in type 2 diabetes. RESULTS AGEs, total sRAGE, cRAGE and the AGEs/sRAGE and AGEs/esRAGE ratios were significantly increased in patients with type 2 diabetes compared to CTR (p < 0.001). In CTR subjects, but not in type 2 diabetes patients, a significant negative correlation between cRAGE and age was confirmed (p = 0.003), whereas the AGEs/sRAGE (p = 0.032) and AGEs/cRAGE (p = 0.006) ratios were positively associated with age. At an average follow-up of 15 years (4,982 person-years), 130 deaths were observed. The increase in the AGEs/cRAGE ratio was accompanied by a higher risk of all-cause mortality in patients with type 2 diabetes (HR per each SD increment = 1.30, 95% CI 1.15-1.47; p < 0.001). Moreover, sRAGE was associated with the development of major adverse cardiovascular events (MACE) in type 2 diabetes patients without previous MACE (OR for each SD increase: 1.48, 95% CI 1.11-1.89). A nomogram based on age, sex, HbA1c, systolic blood pressure, and the AGEs/cRAGE ratio was built to predict 5-, 10- and 15-year survival in type 2 diabetes. Patients were categorized into quartiles of the monogram scores and Kaplan-Meier survival curves confirmed the prognostic accuracy of the model (log-rank p = 6.5 × 10- 13). CONCLUSIONS The ratio between AGEs and the cRAGE isoform is predictive of 15-year survival in patients with type 2 diabetes. Our data support the assessment of circulating AGEs and soluble RAGE isoforms in patients with type 2 diabetes as predictors of MACE and all-cause mortality.
Collapse
Affiliation(s)
- Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy
- Laboratory Medicine Unit, Azienda Ospedaliero Universitaria "Ospedali Riuniti", Ancona, Italy
| | - Stefania Castiglione
- Experimental Cardio-Oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Federica Macrì
- Experimental Cardio-Oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy.
| | - Deborah Ramini
- Clinical Laboratory and Molecular Diagnostic, IRCCS INRCA, Ancona, Italy
| | - Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Elena Tortato
- Metabolic Diseases and Diabetology Department, IRCCS INRCA, Ancona, Italy
| | | | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy
- Clinical Laboratory and Molecular Diagnostic, IRCCS INRCA, Ancona, Italy
| | - Angela Raucci
- Experimental Cardio-Oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| |
Collapse
|
9
|
Normalizing HIF-1α Signaling Improves Cellular Glucose Metabolism and Blocks the Pathological Pathways of Hyperglycemic Damage. Biomedicines 2021; 9:biomedicines9091139. [PMID: 34572324 PMCID: PMC8471680 DOI: 10.3390/biomedicines9091139] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
Intracellular metabolism of excess glucose induces mitochondrial dysfunction and diversion of glycolytic intermediates into branch pathways, leading to cell injury and inflammation. Hyperglycemia-driven overproduction of mitochondrial superoxide was thought to be the initiator of these biochemical changes, but accumulating evidence indicates that mitochondrial superoxide generation is dispensable for diabetic complications development. Here we tested the hypothesis that hypoxia inducible factor (HIF)-1α and related bioenergetic changes (Warburg effect) play an initiating role in glucotoxicity. By using human endothelial cells and macrophages, we demonstrate that high glucose (HG) induces HIF-1α activity and a switch from oxidative metabolism to glycolysis and its principal branches. HIF1-α silencing, the carbonyl-trapping and anti-glycating agent ʟ-carnosine, and the glyoxalase-1 inducer trans-resveratrol reversed HG-induced bioenergetics/biochemical changes and endothelial-monocyte cell inflammation, pointing to methylglyoxal (MGO) as the non-hypoxic stimulus for HIF1-α induction. Consistently, MGO mimicked the effects of HG on HIF-1α induction and was able to induce a switch from oxidative metabolism to glycolysis. Mechanistically, methylglyoxal causes HIF1-α stabilization by inhibiting prolyl 4-hydroxylase domain 2 enzyme activity through post-translational glycation. These findings introduce a paradigm shift in the pathogenesis and prevention of diabetic complications by identifying HIF-1α as essential mediator of glucotoxicity, targetable with carbonyl-trapping agents and glyoxalase-1 inducers.
Collapse
|
10
|
Scavello F, Tedesco CC, Castiglione S, Maciag A, Sangalli E, Veglia F, Spinetti G, Puca AA, Raucci A. Modulation of soluble receptor for advanced glycation end products isoforms and advanced glycation end products in long-living individuals. Biomark Med 2021; 15:785-796. [PMID: 34236256 DOI: 10.2217/bmm-2020-0856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/15/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Circulating levels of soluble receptor for advanced glycation end products (sRAGE) and advanced glycation end products (AGEs) correlate with aging/cardiovascular risk, which is delayed in long-living individuals (LLIs). AGEs/sRAGE isoforms (cleaved RAGE [cRAGE] and secretory RAGE [esRAGE]) ratio is a valuable marker for disease risk. Results: We evaluated circulating sRAGE isoforms, and AGEs in LLIs (n = 95; 90-105 years) and controls (n = 94; 11-89 years). cRAGE decreased with age in controls and further declined in LLIs. esRAGE increased in LLIs. AGEs rose with age in controls and decreased in LLIs that were characterized by a lower AGEs/sRAGE ratio. Notably, cRAGE and AGE/esRAGE ratio better discriminated controls from LLIs. Conclusion: circulating cRAGE could be considered a reliable marker of chronological age while esRAGE a protective factor for longevity.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology & Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Calogero C Tedesco
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology & Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Anna Maciag
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
| | - Elena Sangalli
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Gaia Spinetti
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
| | - Annibale A Puca
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
- Department of Medicine, Surgery & Dentistry, 'Scuola Medica Salernitana', University of Salerno, Baronissi, 84081, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology & Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| |
Collapse
|
11
|
Zhuang A, Yap FYT, Borg DJ, McCarthy D, Fotheringham A, Leung S, Penfold SA, Sourris KC, Coughlan MT, Schulz BL, Forbes JM. The AGE receptor, OST48 drives podocyte foot process effacement and basement membrane expansion (alters structural composition). Endocrinol Diabetes Metab 2021; 4:e00278. [PMID: 34277994 PMCID: PMC8279619 DOI: 10.1002/edm2.278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/16/2021] [Accepted: 05/22/2021] [Indexed: 11/17/2022] Open
Abstract
AIMS The accumulation of advanced glycation end products is implicated in the development and progression of diabetic kidney disease. No study has examined whether stimulating advanced glycation clearance via receptor manipulation is reno-protective in diabetes. Podocytes, which are early contributors to diabetic kidney disease and could be a target for reno-protection. MATERIALS AND METHODS To examine the effects of increased podocyte oligosaccharyltransferase-48 on kidney function, glomerular sclerosis, tubulointerstitial fibrosis and proteome (PXD011434), we generated a mouse with increased oligosaccharyltransferase-48kDa subunit abundance in podocytes driven by the podocin promoter. RESULTS Despite increased urinary clearance of advanced glycation end products, we observed a decline in renal function, significant glomerular damage including glomerulosclerosis, collagen IV deposition, glomerular basement membrane thickening and foot process effacement and tubulointerstitial fibrosis. Analysis of isolated glomeruli identified enrichment in proteins associated with collagen deposition, endoplasmic reticulum stress and oxidative stress. Ultra-resolution microscopy of podocytes revealed denudation of foot processes where there was co-localization of oligosaccharyltransferase-48kDa subunit and advanced glycation end-products. CONCLUSIONS These studies indicate that increased podocyte expression of oligosaccharyltransferase-48 kDa subunit results in glomerular endoplasmic reticulum stress and a decline in kidney function.
Collapse
Affiliation(s)
- Aowen Zhuang
- Glycation and Diabetes ComplicationsMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQldAustralia
- Faculty of MedicineUniversity of QueenslandSt LuciaQldAustralia
- Baker Heart and Diabetes InstituteMelbourneVicAustralia
| | | | - Danielle J. Borg
- Glycation and Diabetes ComplicationsMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQldAustralia
| | - Domenica McCarthy
- Glycation and Diabetes ComplicationsMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQldAustralia
| | - Amelia Fotheringham
- Glycation and Diabetes ComplicationsMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQldAustralia
| | - Sherman Leung
- Glycation and Diabetes ComplicationsMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQldAustralia
| | | | - Karly C. Sourris
- Baker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of DiabetesCentral Clinical SchoolMonash UniversityMelbourneVicAustralia
| | - Melinda T. Coughlan
- Baker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of DiabetesCentral Clinical SchoolMonash UniversityMelbourneVicAustralia
| | - Benjamin L. Schulz
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandSt LuciaQldAustralia
| | - Josephine M. Forbes
- Glycation and Diabetes ComplicationsMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQldAustralia
- Faculty of MedicineUniversity of QueenslandSt LuciaQldAustralia
| |
Collapse
|
12
|
Scavello F, Zeni F, Milano G, Macrì F, Castiglione S, Zuccolo E, Scopece A, Pezone G, Tedesco CC, Nigro P, Degani G, Gambini E, Veglia F, Popolo L, Pompilio G, Colombo GI, Bianchi ME, Raucci A. Soluble Receptor for Advanced Glycation End-products regulates age-associated Cardiac Fibrosis. Int J Biol Sci 2021; 17:2399-2416. [PMID: 34326683 PMCID: PMC8315019 DOI: 10.7150/ijbs.56379] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
Myocardial aging increases the cardiovascular risk in the elderly. The Receptor for Advanced Glycation End-products (RAGE) is involved in age-related disorders. The soluble isoform (sRAGE) acts as a scavenger blocking the membrane-bound receptor activation. This study aims at investigating RAGE contribution to age-related cardiac remodeling. We analyzed the cardiac function of three different age groups of female Rage-/- and C57BL/6N (WT) mice: 2.5- (Young), 12- (Middle-age, MA) and 21-months (Old) old. While aging, Rage-/- mice displayed an increase in left ventricle (LV) dimensions compared to age-matched WT animals, with the main differences observed in the MA groups. Rage-/- mice showed higher fibrosis and a larger number of α-Smooth Muscle Actin (SMA)+ cells with age, along with increased expression of pro-fibrotic Transforming Growth Factor (TGF)-β1 pathway components. RAGE isoforms were undetectable in LV of WT mice, nevertheless, circulating sRAGE declined with aging and inversely associated with LV diastolic dimensions. Human cardiac fibroblasts stimulated with sRAGE exhibited a reduction in proliferation, pro-fibrotic proteins and TGF-beta Receptor 1 (TGFbR1) expression and Smad2-3 activation. Finally, sRAGE administration to MA WT animals reduced cardiac fibrosis. Hence, our work shows that RAGE associates with age-dependent myocardial changes and indicates sRAGE as an inhibitor of cardiac fibroblasts differentiation and age-dependent cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giuseppina Milano
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Federica Macrì
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Estella Zuccolo
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Alessandro Scopece
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giovanni Pezone
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | - Patrizia Nigro
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Genny Degani
- Department of Biosciences, University of Milan, Milan, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Laura Popolo
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Gualtiero I. Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Marco E. Bianchi
- Chromatin Dynamics Unit, San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| |
Collapse
|
13
|
Panizo S, Martínez-Arias L, Alonso-Montes C, Cannata P, Martín-Carro B, Fernández-Martín JL, Naves-Díaz M, Carrillo-López N, Cannata-Andía JB. Fibrosis in Chronic Kidney Disease: Pathogenesis and Consequences. Int J Mol Sci 2021; 22:E408. [PMID: 33401711 PMCID: PMC7795409 DOI: 10.3390/ijms22010408] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a process characterized by an excessive accumulation of the extracellular matrix as a response to different types of tissue injuries, which leads to organ dysfunction. The process can be initiated by multiple and different stimuli and pathogenic factors which trigger the cascade of reparation converging in molecular signals responsible of initiating and driving fibrosis. Though fibrosis can play a defensive role, in several circumstances at a certain stage, it can progressively become an uncontrolled irreversible and self-maintained process, named pathological fibrosis. Several systems, molecules and responses involved in the pathogenesis of the pathological fibrosis of chronic kidney disease (CKD) will be discussed in this review, putting special attention on inflammation, renin-angiotensin system (RAS), parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), Klotho, microRNAs (miRs), and the vitamin D hormonal system. All of them are key factors of the core and regulatory pathways which drive fibrosis, having a great negative kidney and cardiac impact in CKD.
Collapse
Affiliation(s)
- Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Pablo Cannata
- Pathology Department, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Retic REDinREN-ISCIII, 28040 Madrid, Spain;
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - José L. Fernández-Martín
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Jorge B. Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| |
Collapse
|
14
|
Lai CH, Chou CC, Chuang HC, Lin GJ, Pan CH, Chen WL. Receptor for advanced glycation end products in relation to exposure to metal fumes and polycyclic aromatic hydrocarbon in shipyard welders. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 202:110920. [PMID: 32800255 DOI: 10.1016/j.ecoenv.2020.110920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 06/11/2023]
Abstract
Advanced glycation end products (AGE) and the receptor for AGE (RAGE) have been found to be pivotal biomarkers to predict the risk of inflammation and oxidative stress. Limited evidence focuses on the influence of occupational exposure to polycyclic aromatic hydrocarbon (PAH) and metal fumes on AGE and RAGE in shipyard welders. Our aim was to determine the relationships among PAH, metal exposure, and inflammatory biomarkers. From September 1 to December 31, 2017, 53 welding workers (exposed group) and 29 office workers (control group) were enrolled in the study. Comprehensive workups included demographic characteristics, laboratory data, AGE, RAGE, Interleukin-6 (IL-6), tumor necrosis factor-α, PAH, and urinary metal concentrations. RAGE levels were measured by flow cytometric analysis. Urinary 1-hydroxypyrene (1-OHP) was used as a biomarker of exposure to PAH. Several metals were elevated in the personal fine particulate matter (PM2.5) samples, including Mn, Fe, V, Co, Zn, and Cu. The exposed group had significantly higher exposure to PM2.5 (p = 0.015), RAGE (p = 0.020), IL-6 (p = 0.008) than the control group. After adjusting for pertinent variables, there was still a significant and positive association between Ni level and AGE (β = 0.101; 95% CI, 0.031-0.172). Significant relationship between Cr and Cd levels and RAGE was observed (β = 0.173; 95% CI, 0.017-0.329; β = 0.084; 95% CI, 0.011-0.157, respectively). Participants with elevated 1-OHP level had higher odds of high RAGE level in the model 1 (OR = 3.466, 95% CI, 1.053-11.412) and model 2 (OR = 3.454, 95% CI, 1.034-11.536). The RAGE expression of participants was significantly associated with IL-6 levels in the fully adjusted model (β = 0.294; 95% CI, 0.083-0.732). Our findings highlighted that urinary metal levels and PAH were associated with increased AGE and RAGE formation in shipyard workers. Elevated serum RAGE might induce the production of proinflammatory cytokines and trigger ensuing inflammatory cascades.
Collapse
Affiliation(s)
- Ching-Huang Lai
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Chi Chou
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Gu-Jiun Lin
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Hong Pan
- Institute of Labor, Occupational Safety and Health, Ministry of Labor, New Taipei City, Taiwan
| | - Wei-Liang Chen
- Division of Environmental Health & Occupational Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, And School of Medicine, National Defense Medical Center, Taipei, Taiwan; Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, And School of Medicine, National Defense Medical Center, Taipei, Taiwan; Department of Biochemistry, National Defense Medical Center, Taiwan.
| |
Collapse
|
15
|
Pathomthongtaweechai N, Chutipongtanate S. AGE/RAGE signaling-mediated endoplasmic reticulum stress and future prospects in non-coding RNA therapeutics for diabetic nephropathy. Biomed Pharmacother 2020; 131:110655. [PMID: 32853909 DOI: 10.1016/j.biopha.2020.110655] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/01/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
Disturbance of endoplasmic reticulum (ER) homeostasis triggered by the accumulation of unfolded proteins and advanced glycation end-products (AGEs) plays a major role in pathophysiology of diabetic nephropathy. Activation of receptor for AGEs (RAGE) stimulates NADPH oxidase-mediated reactive oxygen species (ROS) production, leading to ER stress, inflammation, glomerular hypertrophy, podocyte injury, and renal fibrosis. A growing body of evidence indicates that non-coding RNAs (ncRNAs) could rescue ER stress and renal inflammation by the epigenetic modification. This review summarizes ncRNA regulation in AGE/RAGE signaling-mediated ER stress, and discusses the opportunities and challenges of ncRNA-loaded extracellular vesicle therapy in diabetic nephropathy.
Collapse
Affiliation(s)
- Nutthapoom Pathomthongtaweechai
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand.
| | - Somchai Chutipongtanate
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand; Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
16
|
Sourris KC, Watson A, Jandeleit-Dahm K. Inhibitors of Advanced Glycation End Product (AGE) Formation and Accumulation. Handb Exp Pharmacol 2020; 264:395-423. [PMID: 32809100 DOI: 10.1007/164_2020_391] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A range of chemically different compounds are known to inhibit the formation and accumulation of advanced glycation end products (AGEs) or disrupt associated signalling pathways. There is evidence that some of these agents can provide end-organ protection in chronic diseases including diabetes. Whilst this group of therapeutics are structurally and functionally different and have a range of mechanisms of action, they ultimately reduce the deleterious actions and the tissue burden of advanced glycation end products. To date it remains unclear if this is due to the reduction in tissue AGE levels per se or the modulation of downstream signal pathways. Some of these agents either stimulate antioxidant defence or reduce the formation of reactive oxygen species (ROS), modify lipid profiles and inhibit inflammation. A number of existing treatments for glucose lowering, hypertension and hyperlipidaemia are also known to reduce AGE formation as a by-product of their action. Targeted AGE formation inhibitors or AGE cross-link breakers have been developed and have shown beneficial effects in animal models of diabetic complications as well as other chronic conditions. However, only a few of these agents have progressed to clinical development. The failure of clinical translation highlights the importance of further investigation of the advanced glycation pathway, the diverse actions of agents which interfere with AGE formation, cross-linking or AGE receptor activation and their effect on the development and progression of chronic diseases including diabetic complications. Advanced glycation end products (AGEs) are (1) proteins or lipids that become glycated as a result of exposure to sugars or (2) non-proteinaceous oxidised lipids. They are implicated in ageing and the development, or worsening, of many degenerative diseases, such as diabetes, atherosclerosis, chronic kidney and Alzheimer's disease. Several antihypertensive and antidiabetic agents and statins also indirectly lower AGEs. Direct AGE inhibitors currently investigated include pyridoxamine and epalrestat, the inhibition of the formation of reactive dicarbonyls such as methylglyoxal as an important precursor of AGEs via increased activation of the detoxifying enzyme Glo-1 and inhibitors of NOX-derived ROS to reduce the AGE/RAGE signalling.
Collapse
Affiliation(s)
- Karly C Sourris
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anna Watson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Karin Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
17
|
Effect of Quamoclit angulata Extract Supplementation on Oxidative Stress and Inflammation on Hyperglycemia-Induced Renal Damage in Type 2 Diabetic Mice. Antioxidants (Basel) 2020; 9:antiox9060459. [PMID: 32471242 PMCID: PMC7346142 DOI: 10.3390/antiox9060459] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is caused by abnormalities of controlling blood glucose and insulin homeostasis. Especially, hyperglycemia causes hyper-inflammation through activation of NLRP3 inflammasome, which can lead to cell apoptosis, hypertrophy, and fibrosis. Quamoclit angulata (QA), one of the annual winders, has been shown ameliorative effects on diabetes. The current study investigated whether the QA extract (QAE) attenuated hyperglycemia-induced renal inflammation related to NLRP inflammasome and oxidative stress in high fat diet (HFD)-induced diabetic mice. After T2DM was induced, the mice were treated with QAE (5 or 10 mg/kg/day) by gavage for 12 weeks. The QAE supplementation reduced homeostasis model assessment insulin resistance (HOMA-IR), kidney malfunction, and glomerular hypertrophy in T2DM. Moreover, the QAE treatment significantly attenuated renal NLRP3 inflammasome dependent hyper-inflammation and consequential renal damage caused by oxidative stress, apoptosis, and fibrosis in T2DM. Furthermore, QAE normalized aberrant energy metabolism (downregulation of p-AMPK, sirtuin (SIRT)-1, and PPARγ-coactivator α (PGC-1 α)) in T2DM mice. Taken together, the results suggested that QAE as a natural product has ameliorative effects on renal damage by regulation of oxidative stress and inflammation in T2DM.
Collapse
|
18
|
Scavello F, Zeni F, Tedesco CC, Mensà E, Veglia F, Procopio AD, Bonfigli AR, Olivieri F, Raucci A. Modulation of soluble receptor for advanced glycation end-products (RAGE) isoforms and their ligands in healthy aging. Aging (Albany NY) 2020; 11:1648-1663. [PMID: 30903794 PMCID: PMC6461165 DOI: 10.18632/aging.101860] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/06/2019] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation end-products (RAGE) recognizes several ligands involved in inflammatory diseases. Two circulating soluble isoforms exist: esRAGE derived from alternative splicing and cRAGE generated by the membrane-bound RAGE (FL-RAGE) proteolysis. Together, esRAGE and cRAGE constitute sRAGE and function as decoy receptors preventing FL-RAGE/ligands binding. We determined serum concentration of both, esRAGE and cRAGE, and their ligands AGEs, HMGB1 and S100A8/A9 in a healthy population of 169 subjects aged 20-90 years. cRAGE showed a negative (r=-0.375, P<0.0001) while AGEs (r=0.160, P=0.0384) and S100A8/A9 (r=0.207, P=0.0091) a positive correlation with age. esRAGE did not change during aging and inversely correlated with Hemoglobin, ALT, insulin, HOMA index, Waist-Hip ratio (W/H), Waist Circumference (WC) and positively with AGEs. cRAGE exhibited also an inverse correlation with WC, W/H, PAI-1, HMGB1, AGEs and S100A8/A9. Age, W/H, HMGB1, S100A8/A9 and AGEs are independent predictors of cRAGE, whereas W/H and AGEs associate with esRAGE. Treatment of cells with glycated albumin reduced cRAGE production and upregulated FL-RAGE. These results indicate that in a healthy population cRAGE is a biomarker of aging while esRAGE represents a more reliable marker of obesity and insulin resistance. Hence, sRAGE isoforms levels could be differentially associated with age-related diseases risk factors.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | - Emanuela Mensà
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | | | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| |
Collapse
|
19
|
Autophagy and mTOR Pathways Mediate the Potential Renoprotective Effects of Vitamin D on Diabetic Nephropathy. Int J Nephrol 2020; 2020:7941861. [PMID: 32455017 PMCID: PMC7243019 DOI: 10.1155/2020/7941861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Not only is diabetic nephropathy (DN) the most common cause of end-stage renal disease worldwide, but it also increases the risk of mortality up to fourteen times compared to normoalbuminuric diabetic patients. Aim The aim of the current study was the evaluation of the renoprotective effects of vitamin D in DN and the possible interplay between autophagy and mTOR pathways. Materials and Methods Fifty male Wistar albino rats were divided (10/group) into control, DN group, insulin-treated DN group, vitamin D-treated DN group, and combined insulin and vitamin D-treated DN group. Assessments of systolic blood pressure, albuminuria, creatinine clearance, serum glucose, insulin, urea, creatinine, inflammatory cytokines, oxidative stress markers, and rat kidney gene expression of mTOR were performed. Histopathological and immunohistochemical assessments of autophagy marker LC3 in rat kidneys were also performed. Results DN was associated with significant increases in SBP, urinary albumin, serum glucose, urea, creatinine, inflammatory cytokines, MDA, and mTOR gene expression (P < 0.05). However, there was significant decrease in creatinine clearance, serum insulin, GSH, and H score value of LC3 when compared with control group (P < 0.05). The combination of insulin and vitamin D treatment significantly restored DN changes when compared with the other treated groups, except in oxidative stress markers where there was an insignificant difference between the combination-treated and insulin-treated groups (P > 0.05). Conclusion It has been concluded that vitamin D is a potent adjuvant therapy in treatment of DN via downregulation of mTOR gene expression, stimulation of autophagy, and antioxidant, anti-inflammatory, and hypotensive effects.
Collapse
|
20
|
Patel DM, Bose M, Cooper ME. Glucose and Blood Pressure-Dependent Pathways-The Progression of Diabetic Kidney Disease. Int J Mol Sci 2020; 21:ijms21062218. [PMID: 32210089 PMCID: PMC7139394 DOI: 10.3390/ijms21062218] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
The major clinical associations with the progression of diabetic kidney disease (DKD) are glycemic control and systemic hypertension. Recent studies have continued to emphasize vasoactive hormone pathways including aldosterone and endothelin which suggest a key role for vasoconstrictor pathways in promoting renal damage in diabetes. The role of glucose per se remains difficult to define in DKD but appears to involve key intermediates including reactive oxygen species (ROS) and dicarbonyls such as methylglyoxal which activate intracellular pathways to promote fibrosis and inflammation in the kidney. Recent studies have identified a novel molecular interaction between hemodynamic and metabolic pathways which could lead to new treatments for DKD. This should lead to a further improvement in the outlook of DKD building on positive results from RAAS blockade and more recently newer classes of glucose-lowering agents such as SGLT2 inhibitors and GLP1 receptor agonists.
Collapse
Affiliation(s)
- Devang M. Patel
- Department of Diabetes, Monash University Central, Clinical School, Melbourne, VIC 3004, Australia;
- Correspondence: (D.M.P.); (M.E.C.)
| | - Madhura Bose
- Department of Diabetes, Monash University Central, Clinical School, Melbourne, VIC 3004, Australia;
| | - Mark E. Cooper
- Department of Diabetes, Monash University Central, Clinical School, Melbourne, VIC 3004, Australia;
- Department of Endocrinology and Diabetes, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Correspondence: (D.M.P.); (M.E.C.)
| |
Collapse
|
21
|
Lespedeza bicolor Extract Ameliorated Renal Inflammation by Regulation of NLRP3 Inflammasome-Associated Hyperinflammation in Type 2 Diabetic Mice. Antioxidants (Basel) 2020; 9:antiox9020148. [PMID: 32050658 PMCID: PMC7071116 DOI: 10.3390/antiox9020148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by hyperglycemia. The chronic hyperglycemic condition causes hyperinflammation via activation of nucleotide-binding oligomerization domain-like pyrin domain containing receptor 3 (NLRP3) inflammasome and abnormally leads to morphological and functional changes in kidney. A previous study showed a protective effect of Lespedeza bicolor extract (LBE) on endothelial dysfunction induced by methylglyoxal glucotoxicity. We aimed to investigate whether LBE ameliorated renal damage through regulation of NLRP3 inflammasome-dependent hyper-inflammation in T2DM mice. After T2DM induction by a high fat diet and low dose of streptozotocin (30 mg/kg), the mice were administered with different dosages of LBE (100 or 250 mg/kg/day) by gavage for 12 weeks. LBE supplementation ameliorated kidney dysfunction demonstrated by urine albumin-creatinine at a low dose and plasma creatinine, blood urea nitrogen (BUN), and glomerular hypertrophy at a high dose. Furthermore, a high dose of LBE supplementation significantly attenuated renal hyper-inflammation associated with NLRP3 inflammasome and oxidative stress related to nuclear factor erythroid 2-related factor 2 (Nrf-2) in T2DM mice. Meanwhile, a low dose of LBE supplementation up-regulated energy metabolism demonstrated by phosphorylation of adenosine monophosphate kinase (AMPK) and Sirtuin (SIRT)-1 in T2DM mice. In conclusion, the current study suggested that LBE, in particular, at a high dose could be used as a beneficial therapeutic for hyperglycemia-induced renal damage in T2DM.
Collapse
|
22
|
Zhuang A, Yap FYT, McCarthy D, Leung C, Sourris KC, Penfold SA, Thallas-Bonke V, Coughlan MT, Schulz BL, Forbes JM. Globally elevating the AGE clearance receptor, OST48, does not protect against the development of diabetic kidney disease, despite improving insulin secretion. Sci Rep 2019; 9:13664. [PMID: 31541173 PMCID: PMC6754370 DOI: 10.1038/s41598-019-50221-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
The accumulation of advanced glycation end products (AGEs) have been implicated in the development and progression of diabetic kidney disease (DKD). There has been interest in investigating the potential of AGE clearance receptors, such as oligosaccharyltransferase-48 kDa subunit (OST48) to prevent the detrimental effects of excess AGE accumulation seen in the diabetic kidney. Here the objective of the study was to increase the expression of OST48 to examine if this slowed the development of DKD by facilitating the clearance of AGEs. Groups of 8-week-old heterozygous knock-in male mice (n = 9-12/group) over-expressing the gene encoding for OST48, dolichyl-diphosphooligosaccharide-protein glycosyltransferase (DDOST+/-) and litter mate controls were randomised to either (i) no diabetes or (ii) diabetes induced via multiple low-dose streptozotocin and followed for 24 weeks. By the study end, global over expression of OST48 increased glomerular OST48. This facilitated greater renal excretion of AGEs but did not affect circulating or renal AGE concentrations. Diabetes resulted in kidney damage including lower glomerular filtration rate, albuminuria, glomerulosclerosis and tubulointerstitial fibrosis. In diabetic mice, tubulointerstitial fibrosis was further exacerbated by global increases in OST48. There was significantly insulin effectiveness, increased acute insulin secretion, fasting insulin concentrations and AUCinsulin observed during glucose tolerance testing in diabetic mice with global elevations in OST48 when compared to diabetic wild-type littermates. Overall, this study suggested that despite facilitating urinary-renal AGE clearance, there were no benefits observed on kidney functional and structural parameters in diabetes afforded by globally increasing OST48 expression. However, the improvements in insulin secretion seen in diabetic mice with global over-expression of OST48 and their dissociation from effects on kidney function warrant future investigation.
Collapse
Affiliation(s)
- Aowen Zhuang
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia.,School of Medicine, University of Queensland, St Lucia, Australia
| | - Felicia Y T Yap
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,Department of Immunology, Central and Eastern Clinical School, AMREP Precinct, Monash University, Melbourne, Australia
| | - Domenica McCarthy
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Chris Leung
- Department of Medicine, University of Melbourne, Austin Hospital, Heidelberg, Australia
| | - Karly C Sourris
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Sally A Penfold
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | | | | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Josephine M Forbes
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia. .,School of Medicine, University of Queensland, St Lucia, Australia. .,Mater Clinical School, University of Queensland, St Lucia, Australia.
| |
Collapse
|
23
|
Abstract
Significance: Obesity and type 2 diabetes mellitus are increasing globally. There is also increasing associated complications, such as non-alcoholic fatty liver disease (NAFLD) and vascular complications of diabetes. There is currently no licensed treatment for NAFLD and no recent treatments for diabetic complications. New approaches are required, particularly those addressing mechanism-based risk factors for health decline and disease progression. Recent Advances: Dicarbonyl stress is the abnormal accumulation of reactive dicarbonyl metabolites such as methylglyoxal (MG) leading to cell and tissue dysfunction. It is a potential driver of obesity, diabetes, and related complications that are unaddressed by current treatments. Increased formation of MG is linked to increased glyceroneogenesis and hyperglycemia in obesity and diabetes and also down-regulation of glyoxalase 1 (Glo1)-which provides the main enzymatic detoxification of MG. Glo1 functional genomics studies suggest that increasing Glo1 expression and activity alleviates dicarbonyl stress; slows development of obesity, related insulin resistance; and prevents development of diabetic nephropathy and other microvascular complications of diabetes. A new therapeutic approach constitutes small-molecule inducers of Glo1 expression-Glo1 inducers-exploiting a regulatory antioxidant response element in the GLO1 gene. A prototype Glo1 inducer, trans-resveratrol (tRES)-hesperetin (HESP) combination, in corrected insulin resistance, improved glycemic control and vascular inflammation in healthy overweight and obese subjects in clinical trial. Critical Issues: tRES and HESP synergize pharmacologically, and HESP likely overcomes the low bioavailability of tRES by inhibition of intestinal glucuronosyltransferases. Future Directions: Glo1 inducers may now be evaluated in Phase 2 clinical trials for treatment of NAFLD and vascular complications of diabetes.
Collapse
Affiliation(s)
- Naila Rabbani
- 1 Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital , Coventry, United Kingdom .,2 Warwick Systems Biology Centre, Senate House, University of Warwick , Coventry, United Kingdom
| | - Paul J Thornalley
- 1 Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital , Coventry, United Kingdom .,2 Warwick Systems Biology Centre, Senate House, University of Warwick , Coventry, United Kingdom
| |
Collapse
|
24
|
Advanced Glycation End Products Stimulate Angiotensinogen Production in Renal Proximal Tubular Cells. Am J Med Sci 2018; 357:57-66. [PMID: 30466736 DOI: 10.1016/j.amjms.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Elevated advanced glycation end products (AGE) in diabetes mellitus (DM) are implicated in the progression of DM-associated tissue injury, including diabetic nephropathy. The intrarenal renin-angiotensin system, in particular augmentation of angiotensinogen (AGT) in proximal tubular cells (PTC), plays a crucial role in the development of diabetic nephropathy. This study investigated hypothesis that AGE stimulates AGT production in PTC. MATERIALS AND METHODS Urinary AGT and AGE levels in streptozotocin-induced DM mice were measured by enzyme-linked immunosorbent assays. AGT expression and secretion were evaluated in cultured rat PTC receiving 0-200 µg/ml AGE-BSA treatments for 24 hours. Furthermore, intracellular signaling pathways activated by AGE were elucidated. RESULTS DM mice exhibited greater urinary AGT and AGE levels compared to control mice (AGT: 21.6 ± 5.5 ng/day vs. 190.1 ± 57.8 ng/day, AGE: 139.1 ± 21.6 μg/day vs. 332.8 ± 102.7 μg/day). In cultured PTC, treatment with AGE-BSA enhanced AGT mRNA expression (3.43 ± 0.11-fold compared to control), intracellular AGT protein levels (3.60 ± 0.38-fold), and secreted AGT levels (2.11 ± 0.18-fold). On the other hand, AGT levels were not altered in PTC receiving nonglycated BSA. Recombinant soluble AGE receptor, which competes with endogenous AGE receptor, diminished the AGE-induced AGT upregulation, suggesting that AGE-BSA stimulates AGT expression via activation of the AGE receptor. Enhanced phosphorylation of ERK1/2 and c-Jun, but not p38 MAP kinase, were observed in AGE-BSA-treated PTC. AGE-induced AGT augmentation was attenuated by an ERK inhibitor. CONCLUSIONS The findings indicate that AGE enhances proximal tubular AGT expression via ERK1/2, which can exacerbate the development of diabetic related kidney injury.
Collapse
|
25
|
Yang DB, Dong XQ, Du Q, Yu WH, Zheng YK, Hu W, Wang KY, Chen FH, Xu YS, Wang Y, Chen G. Clinical relevance of cleaved RAGE plasma levels as a biomarker of disease severity and functional outcome in aneurysmal subarachnoid hemorrhage. Clin Chim Acta 2018; 486:335-340. [PMID: 30144440 DOI: 10.1016/j.cca.2018.08.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cleaved receptor for advanced glycation end-products (cRAGE) has been introduced as a new inflammatory marker. We clarified the associations between cRAGE levels, disease severity and functional outcome in aneurysmal subarachnoid hemorrhage (aSAH). METHODS In this prospective, observational study, plasma levels of total soluble RAGE (sRAGE) and endogenous secretory RAGE (esRAGE) were quantified in 108 aSAH patients and 108 controls. The level of cRAGE was calculated by subtracting the level of esRAGE from that of sRAGE. World Federation of Neurological Surgeons (WFNS) score, modified Fisher score, and Hunt Hess (HH) score were recorded to assess aSAH severity. Relationship between plasma cRAGE levels and 6-month poor outcome (Glasgow Outcome Scale score of 1-3) was assess using multivariate analysis. RESULTS Plasma cRAGE levels were significantly higher in patients than in controls. Its levels were significantly correlated with WNFS score, modified Fisher score and HH score of patients. Plasma cRAGE emerged as an independent predictor for 6-month poor outcome. Area under receiver operating characteristic curve (AUC) of this biomarker was similar to those of WNFS score, modified Fisher score and HH score. Moreover, it significantly improved AUCs of WNFS score, modified Fisher score and HH score. CONCLUSIONS Plasma cRAGE levels are highly associated with the severity and poor prognosis in aSAH.
Collapse
Affiliation(s)
- Ding-Bo Yang
- Department of Neurosurgery, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Xiao-Qiao Dong
- Department of Neurosurgery, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Quan Du
- Department of Neurosurgery, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Wen-Hua Yu
- Department of Neurosurgery, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Yong-Ke Zheng
- Department of Intensive Care Unit, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Wei Hu
- Department of Intensive Care Unit, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Ke-Yi Wang
- Clinical Laboratory Center, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Fang-Hui Chen
- Department of Emergency Medicine, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Yuan-Sheng Xu
- Department of Emergency Medicine, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Yi Wang
- Department of Emergency Medicine, The Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 261 Huansha Road, Hangzhou 310006, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China.
| |
Collapse
|
26
|
Manigrasso MB, Friedman RA, Ramasamy R, D'Agati V, Schmidt AM. Deletion of the formin Diaph1 protects from structural and functional abnormalities in the murine diabetic kidney. Am J Physiol Renal Physiol 2018; 315:F1601-F1612. [PMID: 30132346 DOI: 10.1152/ajprenal.00075.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Diaphanous 1 (DIAPH1), a member of the formin family, binds to the cytoplasmic domain of the receptor for advanced glycation end products (RAGE) and is required for RAGE signal transduction. Experiments employing genetic overexpression or deletion of Ager (the gene encoding RAGE) or its pharmacological antagonism implicate RAGE in the pathogenesis of diabetes-associated nephropathy. We hypothesized that DIAPH1 contributes to pathological and functional derangements in the kidneys of diabetic mice. We show that DIAPH1 is expressed in the human and murine diabetic kidney, at least in part in the tubulointerstitium and glomerular epithelial cells or podocytes. To test the premise that DIAPH1 is linked to diabetes-associated derangements in the kidney, we rendered male mice globally devoid of Diaph1 ( Diaph1-/-) or wild-type controls (C57BL/6 background) diabetic with streptozotocin. Control mice received equal volumes of citrate buffer. After 6 mo of hyperglycemia, diabetic Diaph1-/- mice displayed significantly reduced mesangial sclerosis, podocyte effacement, glomerular basement thickening, and urinary albumin-to-creatinine ratio compared with diabetic mice expressing Diaph1. Analysis of whole kidney cortex revealed that deletion of Diaph1 in diabetic mice significantly reduced expression of genes linked to fibrosis and inflammation. In glomerular isolates, expression of two genes linked to podocyte stress, growth arrest-specific 1 ( Gas1) and cluster of differentiation 36 ( Cd36), was significantly attenuated in diabetic Diaph1-/- mice compared with controls, in parallel with significantly higher levels of nestin (Nes) mRNA, a podocyte marker. Collectively, these data implicate DIAPH1 in the pathogenesis of diabetes-associated nephropathy and suggest that the RAGE-DIAPH1 axis is a logical target for therapeutic intervention in this disorder.
Collapse
Affiliation(s)
- Michaele B Manigrasso
- Diabetes Research Program, Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine , New York, New York
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center, and Department of Biomedical Informatics, Columbia University Irving Medical Center , New York, New York
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine , New York, New York
| | - Vivette D'Agati
- Department of Pathology, College of Physicians and Surgeons, Columbia University , New York, New York
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine , New York, New York
| |
Collapse
|
27
|
Hong J, Wang X, Zhang N, Fu H, Li W. D-ribose induces nephropathy through RAGE-dependent NF-κB inflammation. Arch Pharm Res 2018; 41:838-847. [PMID: 30101366 PMCID: PMC6132794 DOI: 10.1007/s12272-018-1061-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022]
Abstract
Recently, aberrantly high levels of d-ribose have been discovered in type II diabetic patients. d-ribose glycates proteins more rapidly than d-glucose, resulting in the production of advanced glycation end products (AGEs). Accumulations of these products can be found in impaired renal function, but the mechanisms are poorly understood. The present study tested whether d-ribose induces renal dysfunction via the RAGE-dependent NF-κB signaling pathway. In vivo, administration of d-ribose was found to lower blood glucose and regulate insulin tolerance. Compared to controls, urine nitrogen and creatinine excretion were increased in mice receiving d-ribose and were accompanied by severe pathological renal damage. Furthermore, immunohistochemistry showed that NF-κB, AGEs, and receptor of AGEs (RAGE) increased in the kidneys of the mice with d-ribose treatment. In vitro, by western blot and immunofluorescent staining, we confirmed that d-ribose induced NF-κB activation and accumulation of AGEs and RAGE in mesangial cells. By co-immunoprecipitation, we found that the pull-down of RAGE remarkably increased the expression of NF-κB. Silencing the RAGE gene blocked the phosphorylation of NF-κB induced by d-ribose. These results strongly suggest that d-ribose induced NF-κB inflammation in a RAGE-dependent manner, which may be a triggering mechanism leading to nephropathy.
Collapse
Affiliation(s)
- Jinni Hong
- Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Xuemei Wang
- Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Ning Zhang
- Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Hong Fu
- Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Weiwei Li
- Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| |
Collapse
|
28
|
An X, Zhang L, Yao Q, Li L, Wang B, Zhang J, He M, Zhang J. The receptor for advanced glycation endproducts mediates podocyte heparanase expression through NF-κB signaling pathway. Mol Cell Endocrinol 2018; 470:14-25. [PMID: 28478303 DOI: 10.1016/j.mce.2017.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 01/18/2023]
Abstract
Heparanase degrades heparan sulfate in glomerular basement membrane (GBM) and plays an important role in diabetic nephropathy (DN). However, its regulating mechanisms remain to be deciphered. Our present study showed that the major advanced glycation endproducts (AGEs), CML-BSA, significantly increased heparanase expression in cultured podocytes and the effect was blocked by the receptor for advanced glycation endproducts (RAGE) knockdown, antibody and antagonist. In addition, NF-κB p65 phosphorylation was elevated and the increased heparanase expression and secretion upon CML-BSA could be attenuated by NF-κB inhibitor PDTC. Mechanistically, CML-BSA activated heparanase promoter through p65 directly binding to its promoter. Furthermore, the in vivo study showed that serum and renal cortex AGEs levels, glomerular p65 phosphorylation and heparanase expression were significantly increased in DN mice. Taken together, our data suggest that AGEs and RAGE interaction increases podocyte heparanase expression by activating NF-κB signal pathway, which is involved in GBM damages of DN.
Collapse
Affiliation(s)
- Xiaofei An
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Qiuming Yao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Ling Li
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Bin Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Jisheng Zhang
- Department of Otorhinolaryngology, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Ming He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China.
| | - Jinan Zhang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China.
| |
Collapse
|
29
|
Advanced glycation end products in the pathogenesis of chronic kidney disease. Kidney Int 2018; 93:803-813. [DOI: 10.1016/j.kint.2017.11.034] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 10/15/2017] [Accepted: 11/09/2017] [Indexed: 12/11/2022]
|
30
|
Goru SK, Kadakol A, Malek V, Pandey A, Sharma N, Gaikwad AB. Diminazene aceturate prevents nephropathy by increasing glomerular ACE2 and AT 2 receptor expression in a rat model of type1 diabetes. Br J Pharmacol 2017; 174:3118-3130. [PMID: 28688122 DOI: 10.1111/bph.13946] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 06/30/2017] [Accepted: 07/05/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE One of the protective actions of angiotensin converting enzyme-2 (ACE2) is the inactivation of angiotensin II. Expression and activity of ACE2 was reduced in glomeruli of diabetic patients and in animal models of diabetes. Recently the potential role of recombinant ACE2 administration in preventing diabetic nephropathy (DN) has been shown. Here we have tested the effects of the ACE2 activator, diminazene aceturate (DIZE), in a model of DN. EXPERIMENTAL APPROACH Male Wistar rats were rendered diabetic using a single dose of streptozotocin (55 mg·kg-1 , i.p.). After 4 weeks, diabetic animals were divided into experimental groups and treated with DIZE, at a low dose (5 mg·kg-1 ·day-1 ), a high dose (15 mg·kg-1 ·day-1 ) and the high dose with of the AT2 receptor antagonist PD123319 (10 mg·kg-1 ·day-1 ). At the end of the treatment , kidneys from all the groups were collected and processed separately for glomerular isolation, protein isolation, mRNA extraction and for immunohistochemical studies. KEY RESULTS Treatment with DIZE restored ACE2 expression in glomeruli and increased expression of AT2 receptors in whole kidney and isolated glomeruli of diabetic animals. DIZE administration reduced angiotensin II levels and increased angiotensin-(1-7) levels in diabetic kidney. However, PD123319 treatment reversed all these actions of DIZE. CONCLUSIONS AND IMPLICATIONS DIZE treatment reduced diabetes-induced renal damage as shown by reduction of fibrosis and apoptosis. These protective actions of DIZE were blocked by the AT2 receptor antagonist. Taken together, these results suggest that DIZE protected against DN through the ACE2/angiotensin-(1-7)/ AT2 receptor axis.
Collapse
Affiliation(s)
- Santosh Kumar Goru
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Almesh Kadakol
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Vajir Malek
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Anuradha Pandey
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
31
|
Angiotensin II type 2 receptor (AT2R) in renal and cardiovascular disease. Clin Sci (Lond) 2017; 130:1307-26. [PMID: 27358027 DOI: 10.1042/cs20160243] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/06/2016] [Indexed: 12/14/2022]
Abstract
Angiotensin II (Ang II) is well-considered to be the principal effector of the renin-angiotensin system (RAS), which binds with strong affinity to the angiotensin II type 1 (AT1R) and type 2 (AT2R) receptor subtype. However, activation of both receptors is likely to stimulate different signalling mechanisms/pathways and produce distinct biological responses. The haemodynamic and non-haemodynamic effects of Ang II, including its ability to regulate blood pressure, maintain water-electrolyte balance and promote vasoconstriction and cellular growth are well-documented to be mediated primarily by the AT1R. However, its biological and functional effects mediated through the AT2R subtype are still poorly understood. Recent studies have emphasized that activation of the AT2R regulates tissue and organ development and provides in certain context a potential counter-regulatory mechanism against AT1R-mediated actions. Thus, this review will focus on providing insights into the biological role of the AT2R, in particular its actions within the renal and cardiovascular system.
Collapse
|
32
|
Microvascular vasodilator properties of the angiotensin II type 2 receptor in a mouse model of type 1 diabetes. Sci Rep 2017; 7:45625. [PMID: 28361992 PMCID: PMC5374544 DOI: 10.1038/srep45625] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 03/01/2017] [Indexed: 12/02/2022] Open
Abstract
Diabetes Mellitus is associated with severe cardiovascular disorders involving the renin-angiotensin system, mainly through activation of the angiotensin II type 1 receptor (AT1R). Although the type 2 receptor (AT2R) opposes the effects of AT1R, with vasodilator and anti-trophic properties, its role in diabetes is debatable. Thus we investigated AT2R-mediated dilatation in a model of type 1 diabetes induced by streptozotocin in 5-month-old male mice lacking AT2R (AT2R−/y). Glucose tolerance was reduced and markers of inflammation and oxidative stress (cyclooxygenase-2, gp91phox p22phox and p67phox) were increased in AT2R−/y mice compared to wild-type (WT) animals. Streptozotocin-induced hyperglycaemia was higher in AT2R−/y than in WT mice. Arterial gp91phox and MnSOD expression levels in addition to blood 8-isoprostane and creatinine were further increased in diabetic AT2R−/y mice compared to diabetic WT mice. AT2R-dependent dilatation in both isolated mesenteric resistance arteries and perfused kidneys was greater in diabetic mice than in non-diabetic animals. Thus, in type 1 diabetes, AT2R may reduce glycaemia and display anti-oxidant and/or anti-inflammatory properties in association with greater vasodilatation in mesenteric arteries and in the renal vasculature, a major target of diabetes. Therefore AT2R might represent a new therapeutic target in diabetes.
Collapse
|
33
|
Antonelli A, Di Maggio S, Rejman J, Sanvito F, Rossi A, Catucci A, Gorzanelli A, Bragonzi A, Bianchi ME, Raucci A. The shedding-derived soluble receptor for advanced glycation endproducts sustains inflammation during acute Pseudomonas aeruginosa lung infection. Biochim Biophys Acta Gen Subj 2017; 1861:354-364. [DOI: 10.1016/j.bbagen.2016.11.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 01/07/2023]
|
34
|
High Mobility Group Box-1: A Missing Link between Diabetes and Its Complications. Mediators Inflamm 2016; 2016:3896147. [PMID: 27847406 PMCID: PMC5099456 DOI: 10.1155/2016/3896147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/03/2016] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 (HMGB-1), a damage-associated molecular pattern, can be actively or passively released from various cells under different conditions and plays a pivotal role in the pathogenesis of inflammation and angiogenesis-dependent diseases. More and more evidence suggests that inflammation, in addition to its role in progression of diabetes, also promotes initiation and development of diabetic complications. In this review, we focus on the role of HMGB-1 in diabetes-related complications and the therapeutic strategies targeting HMGB-1 in diabetic complications.
Collapse
|
35
|
Sadar S, Kaspate D, Vyawahare N. Protective effect of L-glutamine against diabetes-induced nephropathy in experimental animal: Role of KIM-1, NGAL, TGF-β1, and collagen-1. Ren Fail 2016; 38:1483-1495. [PMID: 27756197 DOI: 10.1080/0886022x.2016.1227918] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Diabetic nephropathy is a serious microvascular complication and one of the main causes of end-stage renal disease. L-Glutamine (LG) is naturally occurring amino acids with antidiabetic and antioxidant potential. The aim of present investigation was to evaluate the potential of LG against streptozotocin (STZ)-induced diabetic nephropathy (DN) in laboratory rats. DN was induced in male Wistar rats (200-220 g) by intraperitoneal administration of STZ (55 mg/kg). Animals were treated orally with either distilled water (10 mg/kg) or LG (250, 500, and 1000 mg/kg) or Sitagliptin (5 mg/kg). Various biochemical, molecular, and histological (hematoxylin-eosin and Masson's trichrome stain) parameters were assessed. Administration of LG (500 and 1000 mg/kg) significantly inhibited (p < .05) STZ-induced alterations in serum and urine biochemistry (urine creatinine, uric acid, albumin, and BUN). It also significantly increased creatinine clearance rate. STZ induced increase in renal oxidonitrosative stress was significantly decreased (p < .05) by LG (500 and 1000 mg/kg) treatment. Upregulated renal KIM-1, NGAL, TGF-β1, and collagen-1 mRNA expression after STZ administration was significantly inhibited (p < .05) by LG (500 and 1000 mg/kg) treatment. Correlation analysis also revealed that antidiabetic potential of LG attenuates STZ-induced elevated renal KIM-1, NGAL, TGF-β1, and collagen-1 mRNA expression. Histopathological alteration induced by STZ in renal tissue was ameliorated by LG treatment. In conclusion, results of present investigation suggest that treatment with LG ameliorated STZ-induced DN via the inhibition of oxidonitrosative stress as well as downregulation of KIM-1, NGAL, TGF-β1, and collagen-1 mRNA expressions.
Collapse
Affiliation(s)
- Smeeta Sadar
- a Padmashree Dr D. Y. Patil College of Pharmacy , Akurdi , Pune , Maharashtra , India
| | - Dipti Kaspate
- b Cognizant Technology Solution , Hinjewadi, Pune , Maharashtra , India
| | - Neeraj Vyawahare
- a Padmashree Dr D. Y. Patil College of Pharmacy , Akurdi , Pune , Maharashtra , India
| |
Collapse
|
36
|
Di Maggio S, Gatti E, Liu J, Bertolotti M, Fritz G, Bianchi ME, Raucci A. The Mouse-Specific Splice Variant mRAGE_v4 Encodes a Membrane-Bound RAGE That Is Resistant to Shedding and Does Not Contribute to the Production of Soluble RAGE. PLoS One 2016; 11:e0153832. [PMID: 27655137 PMCID: PMC5031469 DOI: 10.1371/journal.pone.0153832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/02/2016] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation end-products (RAGE) is involved in the onset and progression of several inflammatory diseases. The RAGE primary transcript undergoes numerous alternative splicing (AS) events, some of which are species-specific. Here, we characterize the mouse-specific mRAGE_v4 splice variant, which is conserved in rodents and absent in primates. mRAGE_v4 derives from exon 9 skipping and encodes a receptor (M-RAGE) that lacks 9 amino acids between the transmembrane and the immunoglobulin (Ig) domains. RNA-Seq data confirm that in mouse lung mRAGE_v4 is the most abundant RAGE mRNA isoform after mRAGE, which codes for full-length RAGE (FL-RAGE), while in heart all RAGE variants are almost undetectable. The proteins M-RAGE and FL-RAGE are roughly equally abundant in mouse lung. Contrary to FL-RAGE, M-RAGE is extremely resistant to shedding because it lacks the peptide motif recognized by both ADAM10 and MMP9, and does not contribute significantly to soluble cRAGE formation. Thus, a cassette exon in RAGE corresponds to a specific function of the RAGE protein–the ability to be shed. Given the differences in RAGE AS variants between rodents and humans, caution is due in the interpretation of results obtained in mouse models of RAGE-dependent human pathologies.
Collapse
Affiliation(s)
- Stefania Di Maggio
- Experimental Cardio-Oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Elena Gatti
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Jaron Liu
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Matteo Bertolotti
- Experimental Cardio-Oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Günter Fritz
- Institute for Neuropathology, University of Freiburg, Freiburg, Germany
| | - Marco E. Bianchi
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
- Università Vita Salute San Raffaele, Milano, Italy
| | - Angela Raucci
- Experimental Cardio-Oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
- * E-mail:
| |
Collapse
|
37
|
Gray SP, Di Marco E, Kennedy K, Chew P, Okabe J, El-Osta A, Calkin AC, Biessen EA, Touyz RM, Cooper ME, Schmidt HH, Jandeleit-Dahm KA. Reactive Oxygen Species Can Provide Atheroprotection via NOX4-Dependent Inhibition of Inflammation and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2016; 36:295-307. [DOI: 10.1161/atvbaha.115.307012] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 12/18/2015] [Indexed: 02/07/2023]
Abstract
Objective—
Oxidative stress is considered a hallmark of atherosclerosis. In particular, the superoxide-generating type 1 NADPH oxidase (NOX1) has been shown to be induced and play a pivotal role in early phases of mouse models of atherosclerosis and in the context of diabetes mellitus. Here, we investigated the role of the most abundant type 4 isoform (NOX4) in human and mouse advanced atherosclerosis.
Approach and Results—
Plaques of patients with cardiovascular events or established diabetes mellitus showed a surprising reduction in expression of the most abundant but hydrogen peroxide (H
2
O
2
)-generating type 4 isoform (Nox4), whereas Nox1 mRNA was elevated, when compared with respective controls. As these data suggested that NOX4-derived reactive oxygen species may convey a surprisingly protective effect during plaque progression, we examined a mouse model of accelerated and advanced diabetic atherosclerosis, the streptozotocin-treated
ApoE
−/−
mouse, with (
NOX4
−/−
) and without genetic deletion of Nox4. Similar to the human data, advanced versus early plaques of wild-type mice showed reduced Nox4 mRNA expression. Consistent with a rather protective role of NOX4-derived reactive oxygen species,
NOX4
−/−
mice showed increased atherosclerosis when compared with wild-type mice. Deleting NOX4 was associated with reduced H
2
O
2
forming activity and attenuation of the proinflammatory markers, monocyte chemotratic protein-1, interleukin-1β, and tumor necrosis factor-α, as well as vascular macrophage accumulation. Furthermore, there was a greater accumulation of fibrillar collagen fibres within the vascular wall and plaque in diabetic
Nox4
−/−
ApoE
−/−
mice, indicative of plaque remodeling. These data could be replicated in human aortic endothelial cells in vitro, where Nox4 overexpression increased H
2
O
2
and reduced the expression of pro-oxidants and profibrotic markers. Interestingly, Nox4 levels inversely correlated with Nox2 gene and protein levels. Although NOX2 is not constitutively active unlike NOX4 and forms rather superoxide, this opens up the possibility that at least some effects of NOX4 deletion are mediated by NOX2 activation.
Conclusions—
Thus, the appearance of reactive oxygen species in atherosclerosis is apparently not always a nondesirable oxidative stress, but can also have protective effects. Both in humans and in mouse, the H
2
O
2
-forming NOX4, unlike the superoxide-forming NOX1, can act as a negative modulator of inflammation and remodeling and convey atheroprotection. These results have implications on how to judge reactive oxygen species formation in cardiovascular disease and need to be considered in the development of NOX inhibitory drugs.
Collapse
Affiliation(s)
- Stephen P. Gray
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Elyse Di Marco
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Kit Kennedy
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Phyllis Chew
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Jun Okabe
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Assam El-Osta
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Anna C. Calkin
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Erik A.L. Biessen
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Rhian M. Touyz
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Mark E. Cooper
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Harald H.H.W. Schmidt
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| | - Karin A.M. Jandeleit-Dahm
- From the Diabetic Complications Laboratory (S.P.G., E.D.M., K.K., P.C., M.E.C., K.A.M.J.-D.), Epigenetics Laboratory (J.O., A.E.-O.), and Diabetes and Dyslipidaemia Group (A.C.C.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Faculty of Medicine, Monash University, Melbourne, Australia (S.P.G., E.D.M., K.A.M.J.-D.); Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands (E.A.L.B.); Institute of Cardiovascular and Medical Sciences, University of
| |
Collapse
|
38
|
Gallic acid ameliorates renal functions by inhibiting the activation of p38 MAPK in experimentally induced type 2 diabetic rats and cultured rat proximal tubular epithelial cells. Chem Biol Interact 2015; 240:292-303. [PMID: 26341651 DOI: 10.1016/j.cbi.2015.08.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/14/2015] [Accepted: 08/31/2015] [Indexed: 12/26/2022]
Abstract
Diabetic nephropathy (DN) is one of the leading causes of morbidity and mortality in diabetic patients that accounts for about 40% of deaths in type 2 diabetes. p38 mitogen activated protein kinase (p38 MAPK), a serine-threonine kinase, plays an important role in tissue inflammation and is known to be activated under conditions of oxidative stress and hyperglycemia. The role of p38 MAPK has been demonstrated in DN, and its inhibition has been suggested as an alternative approach in the treatment of DN. In the present study, we investigated the nephroprotective effects of an anti-inflammatory phenolic compound, gallic acid (GA, 3,4,5-trihydroxybenzoic acid), in high fat diet/streptozotocin (HFD/STZ) induce type 2 diabetic wistar albino rats. GA (25 mg/kgbw and 50 mg/kgbw, p.o.) treatment for 16 weeks post induction of diabetes led to a significant reduction in the levels of blood glucose, HbA1c, serum creatinine, blood urea nitrogen and proteinuria as well as a significant reduction in the levels of creatinine clearance. GA significantly inhibited the renal p38 MAPK and nuclear factor kappa B (N-κB) activation as well as significantly reduced the levels of renal transforming growth factor beta (TGF-β) and fibronectin. Treatment with GA resulted in a significant reduction in the serum levels of proinflammatory cytokines viz. interleukin 1 beta (IL-1β), IL-6 and tumor necrosis factor alpha (TNF-α). Moreover, GA significantly lowered renal pathology and attenuated renal oxidative stress. In cultured rat NRK 52E proximal tubular epithelial cells, GA treatment inhibited high glucose induced activation of p38 MAPK and NF-κB as well as suppressed proinflammatory cytokine synthesis. The results of the present study provide in vivo and in vitro evidences that the p38 MAPK pathway plays an important role in the pathogenesis of DN, and GA attenuates the p38 MAPK-mediated renal dysfunction in HFD/STZ induced type 2 diabetic rats.
Collapse
|
39
|
Gugliucci A, Menini T. The axis AGE-RAGE-soluble RAGE and oxidative stress in chronic kidney disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 824:191-208. [PMID: 25039001 DOI: 10.1007/978-3-319-07320-0_14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Chronic kidney disease (CKD) has been shown to be associated with high oxidative stress and cardiovascular disease. In this chapter our focus will be on the role of advanced glycation end products (AGE) and their receptor, RAGE in CKD progression and their role on cardiovascular complications. We provide a succinct, yet comprehensive summary of the current knowledge, the challenges and the future therapeutic avenues that are stemming out from novel recent findings. We first briefly review glycation and AGE formation and the role of the kidney in their metabolism. Next, we focus on the RAGE, its signaling and role in oxidative stress. We address the possible role of soluble RAGEs as decoys and the controversy regarding this issue. We then provide the latest information on the specific role of both AGE and RAGE in inflammation and perpetuation of kidney damage in diabetes and in CKD without diabetes, which is the main purpose of the review. Finally, we offer an update on new avenues to target the AGE-RAGE axis in CKD.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Department of Research, College of Osteopathic Medicine, Touro University-California, 1310 Club Drive, 94592, Vallejo, CA, USA,
| | | |
Collapse
|
40
|
Tikellis C, Pickering RJ, Tsorotes D, Huet O, Cooper ME, Jandeleit-Dahm K, Thomas MC. Dicarbonyl stress in the absence of hyperglycemia increases endothelial inflammation and atherogenesis similar to that observed in diabetes. Diabetes 2014; 63:3915-25. [PMID: 24812427 DOI: 10.2337/db13-0932] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The deleterious effects of high glucose levels and enhanced metabolic flux on the vasculature are thought to be mediated by the generation of toxic metabolites, including reactive dicarbonyls like methylglyoxal (MG). In this article, we demonstrate that increasing plasma MG to levels observed in diabetic mice either using an exogenous source (1% in drinking water) or generated following inhibition, its primary clearance enzyme, glyoxalase-1 (with 50 mg/kg IP bromobenzyl-glutathione cyclopentyl diester every second day), was able to increase vascular adhesion and augment atherogenesis in euglycemic apolipoprotein E knockout mice to a similar magnitude as that observed in hyperglycemic mice with diabetes. The effects of MG appear partly mediated by activation of the receptor for advanced glycation end products (RAGE), as deletion of RAGE was able to reduce inflammation and atherogenesis associated with MG exposure. However, RAGE deletion did not completely prevent inflammation or vascular damage, possibly because the induction of mitochondrial oxidative stress by dicarbonyls also contributes to inflammation and atherogenesis. Such data would suggest that a synergistic combination of RAGE antagonism and antioxidants may offer the greatest utility for the prevention and management of diabetic vascular complications.
Collapse
Affiliation(s)
- Chris Tikellis
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia Central Clinical School, Clinical Hematology, Monash University, Melbourne, Australia
| | | | | | - Olivier Huet
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia Intensive Care Unit, The Alfred Hospital, Melbourne, Australia
| | - Mark E Cooper
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia Department of Medicine, Monash University, Melbourne, Australia
| | - Karin Jandeleit-Dahm
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia Department of Medicine, Monash University, Melbourne, Australia
| | - Merlin C Thomas
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
41
|
Tesch G, Sourris KC, Summers SA, McCarthy D, Ward MS, Borg DJ, Gallo LA, Fotheringham AK, Pettit AR, Yap FYT, Harcourt BE, Tan ALY, Kausman JY, Nikolic-Paterson D, Kitching AR, Forbes JM. Deletion of bone-marrow-derived receptor for AGEs (RAGE) improves renal function in an experimental mouse model of diabetes. Diabetologia 2014; 57:1977-85. [PMID: 24957662 DOI: 10.1007/s00125-014-3291-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/09/2014] [Indexed: 01/11/2023]
Abstract
AIMS/HYPOTHESIS The AGEs and the receptor for AGEs (RAGE) are known contributors to diabetic complications. RAGE also has a physiological role in innate and adaptive immunity and is expressed on immune cells. The aim of this study was to determine whether deletion of RAGE from bone-marrow-derived cells influences the pathogenesis of experimental diabetic nephropathy. METHODS Groups (n = 8/group) of lethally irradiated 8 week old wild-type (WT) mice were reconstituted with bone marrow from WT (WT → WT) or RAGE-deficient (RG) mice (RG → WT). Diabetes was induced using multiple low doses of streptozotocin after 8 weeks of bone marrow reconstitution and mice were followed for a further 24 weeks. RESULTS Compared with diabetic WT mice reconstituted with WT bone marrow, diabetic WT mice reconstituted with RG bone marrow had lower urinary albumin excretion and podocyte loss, more normal creatinine clearance and less tubulo-interstitial injury and fibrosis. However, glomerular collagen IV deposition, glomerulosclerosis and cortical levels of TGF-β were not different among diabetic mouse groups. The renal tubulo-interstitium of diabetic RG → WT mice also contained fewer infiltrating CD68(+) macrophages that were activated. Diabetic RG → WT mice had lower renal cortical concentrations of CC chemokine ligand 2 (CCL2), macrophage inhibitory factor (MIF) and IL-6 than diabetic WT → WT mice. Renal cortical RAGE ligands S100 calgranulin (S100A)8/9 and AGEs, but not high mobility box protein B-1 (HMGB-1) were also decreased in diabetic RG → WT compared with diabetic WT → WT mice. In vitro, bone-marrow-derived macrophages from WT but not RG mice stimulated collagen IV production in cultured proximal tubule cells. CONCLUSIONS/INTERPRETATION These studies suggest that RAGE expression on haemopoietically derived immune cells contributes to the functional changes seen in diabetic nephropathy by promoting macrophage infiltration and renal tubulo-interstitial damage.
Collapse
Affiliation(s)
- Greg Tesch
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Melbourne, VIC, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gray SP, Jandeleit-Dahm K. The pathobiology of diabetic vascular complications--cardiovascular and kidney disease. J Mol Med (Berl) 2014; 92:441-52. [PMID: 24687627 DOI: 10.1007/s00109-014-1146-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/03/2014] [Accepted: 03/14/2014] [Indexed: 02/06/2023]
Abstract
With the increasing incidence of obesity and type 2 diabetes, it is predicted that more than half of Americans will have diabetes or pre-diabetes by 2020. Diabetic patients develop vascular complications at a much faster rate in comparison to non-diabetic individuals, and cardiovascular risk is increased up to tenfold. With the increasing incidence of diabetes across the world, the development of vascular complications will become an increasing medical burden. Diabetic vascular complications affect the micro- and macro-vasculature leading to kidney disease often requiring dialysis and transplantation or cardiovascular disease increasing the risk for myocardial infarction, stroke and amputations as well as leading to premature mortality. It has been suggested that many complex pathways contribute to the pathobiology of diabetic complications including hyperglycaemia itself, the production of advanced glycation end products (AGEs) and interaction with the receptors for AGEs such as the receptor for advanced glycation end products (RAGE), as well as the activation of vasoactive systems such as the renin-angiotensin aldosterone system (RAAS) and the endothelin system. More recently, it has been hypothesised that reactive oxygen species derived from NAD(P)H oxidases (Nox) may represent a common downstream mediator of vascular injury in diabetes. Current standard treatment of care includes the optimization of blood glucose and blood pressure usually including inhibitors of the renin-angiotensin system. Although these interventions are able to delay progression, they fail to prevent the development of complications. Thus, there is an urgent medical need to identify novel targets in diabetic vascular complications which may include the blockade of Nox-derived ROS formation, as well as blockade of AGE formation and inhibitors of RAGE activation. These strategies may provide superior protection against the deleterious effects of diabetes on the vasculature.
Collapse
Affiliation(s)
- Stephen P Gray
- Diabetes Complications Division, Baker IDI Heart & Diabetes Research Institute, PO Box 6492, St Kilda Rd, Melbourne, VIC, 8008, Australia,
| | | |
Collapse
|
43
|
Milutinovic PS, Englert JM, Crum LT, Mason NS, Ramsgaard L, Enghild JJ, Sparvero LJ, Lotze MT, Oury TD. Clearance kinetics and matrix binding partners of the receptor for advanced glycation end products. PLoS One 2014; 9:e88259. [PMID: 24642901 PMCID: PMC3958346 DOI: 10.1371/journal.pone.0088259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 01/07/2014] [Indexed: 01/11/2023] Open
Abstract
Elucidating the sites and mechanisms of sRAGE action in the healthy state is vital to better understand the biological importance of the receptor for advanced glycation end products (RAGE). Previous studies in animal models of disease have demonstrated that exogenous sRAGE has an anti-inflammatory effect, which has been reasoned to arise from sequestration of pro-inflammatory ligands away from membrane-bound RAGE isoforms. We show here that sRAGE exhibits in vitro binding with high affinity and reversibly to extracellular matrix components collagen I, collagen IV, and laminin. Soluble RAGE administered intratracheally, intravenously, or intraperitoneally, does not distribute in a specific fashion to any healthy mouse tissue, suggesting against the existence of accessible sRAGE sinks and receptors in the healthy mouse. Intratracheal administration is the only effective means of delivering exogenous sRAGE to the lung, the organ in which RAGE is most highly expressed; clearance of sRAGE from lung does not differ appreciably from that of albumin.
Collapse
Affiliation(s)
- Pavle S. Milutinovic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Judson M. Englert
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Lauren T. Crum
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Neale S. Mason
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Lasse Ramsgaard
- Center for Insoluble Protein Structures, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J. Enghild
- Center for Insoluble Protein Structures, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Louis J. Sparvero
- Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Michael T. Lotze
- Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Tim D. Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
44
|
Park EY, Kim BH, Lee EJ, Chang E, Kim DW, Choi SY, Park JH. Targeting of receptor for advanced glycation end products suppresses cyst growth in polycystic kidney disease. J Biol Chem 2014; 289:9254-62. [PMID: 24515114 DOI: 10.1074/jbc.m113.514166] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited renal disorder. Although a myriad of research groups have attempted to identify a new therapeutic target for ADPKD, no drug has worked well in clinical trials. Our research group has focused on the receptor for advanced glycation end products (RAGE) gene as a novel target for ADPKD. This gene is involved in inflammation and cell proliferation. We have already confirmed that blocking RAGE function attenuates cyst growth in vitro. Based on this previous investigation, our group examined the effect of RAGE on cyst enlargement in vivo. PC2R mice, a severe ADPKD mouse model that we generated, were utilized. An adenovirus containing anti-RAGE shRNA was injected intravenously into this model. We observed that RAGE gene knockdown resulted in loss of kidney weight and volume. Additionally, the cystic area that originated from different nephron segments decreased in size because of down-regulation of the RAGE gene. Blood urea nitrogen and creatinine values tended to be lower after inhibiting RAGE. Based on these results, we confirmed that the RAGE gene could be an effective target for ADPKD treatment.
Collapse
Affiliation(s)
- Eun Young Park
- From the Department of Biological Science, Sookmyung Women's University, Seoul 140-742, Republic of Korea and
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Diabetes mellitus contributes greatly to morbidity, mortality, and overall health care costs. In major part, these outcomes derive from the high incidence of progressive kidney dysfunction in patients with diabetes making diabetic nephropathy a leading cause of end-stage renal disease. A better understanding of the molecular mechanism involved and of the early dysfunctions observed in the diabetic kidney may permit the development of new strategies to prevent diabetic nephropathy. Here we review the pathophysiological changes that occur in the kidney in response to hyperglycemia, including the cellular responses to high glucose and the responses in vascular, glomerular, podocyte, and tubular function. The molecular basis, characteristics, and consequences of the unique growth phenotypes observed in the diabetic kidney, including glomerular structures and tubular segments, are outlined. We delineate mechanisms of early diabetic glomerular hyperfiltration including primary vascular events as well as the primary role of tubular growth, hyperreabsorption, and tubuloglomerular communication as part of a "tubulocentric" concept of early diabetic kidney function. The latter also explains the "salt paradox" of the early diabetic kidney, that is, a unique and inverse relationship between glomerular filtration rate and dietary salt intake. The mechanisms and consequences of the intrarenal activation of the renin-angiotensin system and of diabetes-induced tubular glycogen accumulation are discussed. Moreover, we aim to link the changes that occur early in the diabetic kidney including the growth phenotype, oxidative stress, hypoxia, and formation of advanced glycation end products to mechanisms involved in progressive kidney disease.
Collapse
Affiliation(s)
- Volker Vallon
- Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA.
| | | |
Collapse
|
46
|
Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal 2013; 25:2185-97. [DOI: 10.1016/j.cellsig.2013.06.013] [Citation(s) in RCA: 347] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/15/2013] [Accepted: 06/25/2013] [Indexed: 01/03/2023]
|
47
|
Renal Protective Role of Xiexin Decoction with Multiple Active Ingredients Involves Inhibition of Inflammation through Downregulation of the Nuclear Factor-κB Pathway in Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:715671. [PMID: 23935673 PMCID: PMC3713598 DOI: 10.1155/2013/715671] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/28/2013] [Indexed: 12/01/2022]
Abstract
In Chinese medicine, Xiexin decoction (XXD) has been used for the clinical treatment of diabetes for at least 1700 years. The present study was conducted to investigate the effective ingredients of XXD and their molecular mechanisms of antidiabetic nephropathy in rats. Rats with diabetes induced by high-fat diet and streptozotocin were treated with XXD extract for 12 weeks. XXD significantly improved the glucolipid metabolism disorder, attenuated albuminuria and renal pathological changes, reduced renal advanced glycation end-products, inhibited receptor for advanced glycation end-product and inflammation factors expression, suppressed renal nuclear factor-κB pathway activity, and downregulated renal transforming growth factor-β1. The concentrations of multiple components in plasma from XXD were determined by liquid chromatography and tandem mass spectrometry. Pharmacokinetic/pharmacodynamic analysis using partial least square regression revealed that 8 ingredients of XXD were responsible for renal protective effects via actions on multiple molecular targets. Our study suggests that the renal protective role of XXD with multiple effective ingredients involves inhibition of inflammation through downregulation of the nuclear factor-κB pathway, reducing renal advanced glycation end-products and receptor for advanced glycation end-product in diabetic rats.
Collapse
|
48
|
Gray SP, Di Marco E, Okabe J, Szyndralewiez C, Heitz F, Montezano AC, de Haan JB, Koulis C, El-Osta A, Andrews KL, Chin-Dusting JPF, Touyz RM, Wingler K, Cooper ME, Schmidt HHHW, Jandeleit-Dahm KA. NADPH Oxidase 1 Plays a Key Role in Diabetes Mellitus–Accelerated Atherosclerosis. Circulation 2013; 127:1888-902. [PMID: 23564668 DOI: 10.1161/circulationaha.112.132159] [Citation(s) in RCA: 292] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Stephen P. Gray
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Elyse Di Marco
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Jun Okabe
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Cedric Szyndralewiez
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Freddy Heitz
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Augusto C. Montezano
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Judy B. de Haan
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Christine Koulis
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Assam El-Osta
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Karen L. Andrews
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Jaye P. F. Chin-Dusting
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Rhian M. Touyz
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Kirstin Wingler
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Mark E. Cooper
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Harald H. H. W. Schmidt
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Karin A Jandeleit-Dahm
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| |
Collapse
|
49
|
Thallas-Bonke V, Coughlan MT, Tan ALY, Harcourt BE, Morgan PE, Davies MJ, Bach LA, Cooper ME, Forbes JM. Targeting the AGE-RAGE axis improves renal function in the context of a healthy diet low in advanced glycation end-product content. Nephrology (Carlton) 2012; 18:47-56. [DOI: 10.1111/j.1440-1797.2012.01665.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2012] [Indexed: 01/12/2023]
Affiliation(s)
| | - Melinda T Coughlan
- Diabetes Complications Division; Baker IDI Heart & Diabetes Institute; Melbourne; Victoria; Australia
| | | | | | | | - Michael J Davies
- Free Radical Group; The Heart Research Institute; Sydney; New South Wales; Australia
| | - Leon A Bach
- Department of Medicine and Immunology; AMREP Precinct; Monash University; Melbourne; Victoria; Australia
| | | | | |
Collapse
|
50
|
Watson AMD, Gray SP, Jiaze L, Soro-Paavonen A, Wong B, Cooper ME, Bierhaus A, Pickering R, Tikellis C, Tsorotes D, Thomas MC, Jandeleit-Dahm KAM. Alagebrium reduces glomerular fibrogenesis and inflammation beyond preventing RAGE activation in diabetic apolipoprotein E knockout mice. Diabetes 2012; 61:2105-13. [PMID: 22698914 PMCID: PMC3402321 DOI: 10.2337/db11-1546] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Advanced glycation end products (AGEs) are important mediators of diabetic nephropathy that act through the receptor for AGEs (RAGE), as well as other mechanisms, to promote renal inflammation and glomerulosclerosis. The relative contribution of RAGE-dependent and RAGE-independent signaling pathways has not been previously studied in vivo. In this study, diabetic RAGE apoE double-knockout (KO) mice with streptozotocin-induced diabetes were treated with the AGE inhibitor, alagebrium (1 mg/kg/day), or the ACE inhibitor, quinapril (30 mg/kg/day), for 20 weeks, and renal parameters were assessed. RAGE deletion attenuated mesangial expansion, glomerular matrix accumulation, and renal oxidative stress associated with 20 weeks of diabetes. By contrast, inflammation and AGE accumulation associated with diabetes was not prevented. However, treatment with alagebrium in diabetic RAGE apoE KO mice reduced renal AGE levels and further reduced glomerular matrix accumulation. In addition, even in the absence of RAGE expression, alagebrium attenuated cortical inflammation, as denoted by the reduced expression of monocyte chemoattractant protein-1, intracellular adhesion molecule-1, and the macrophage marker cluster of differentiation molecule 11b. These novel findings confirm the presence of important RAGE-independent as well as RAGE-dependent signaling pathways that may be activated in the kidney by AGEs. This has important implications for the design of optimal therapeutic strategies for the prevention of diabetic nephropathy.
Collapse
Affiliation(s)
- Anna M D Watson
- Diabetes Complications Division, Diabetes and Kidney Disease, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|