1
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Mechanism of ferroptosis in heart failure: The role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and intervention strategies. Ageing Res Rev 2025:102770. [PMID: 40360081 DOI: 10.1016/j.arr.2025.102770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
The ferroptosis of cardiomyocytes has been recognized as the core pathological mechanism of heart failure. During the evolution of cardiovascular diseases, the accumulation of angiotensin II and advanced glycation end products can lead to the excessive activation of the RAGE/TLR4-JNK1/2 pathway, which subsequently triggers ferritinophagy, clockophagy, and enhanced p53 activity, ultimately leading to cardiomyocyte ferroptosis. It is evident that deeply unraveling the specific mechanisms in this field and comprehensively evaluating potential drugs and therapeutic strategies targeting this pathway is crucial for improving the status of cardiomyocyte ferroptosis. However, our current understanding of this pathway's specific molecular biological mechanisms in the process of cardiomyocyte ferroptosis remains limited. In light of this, this paper first comprehensively reviews the historical context of ferroptosis research, compares the similarities and differences between ferroptosis and other standard modes of cell death, elucidates the core mechanisms of ferroptosis and its close connection with heart failure, aiming to establish a basic cognitive framework for readers on ferroptosis and its role in heart failure. Subsequently, the paper delves into the pivotal role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and its intricate molecular biological regulatory network. Furthermore, it systematically integrates various therapeutic approaches aimed at inhibiting RAGE, TLR4, and JNK1/2 activity to alleviate cardiomyocyte ferroptosis, encompassing RNA interference technology, gene knockout techniques, small molecule inhibitors, natural active ingredients, as well as traditional Chinese and Western medicines, with the ultimate goal of forging new avenues and strategies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| |
Collapse
|
2
|
Vianello E, Beltrami AP, Aleksova A, Janjusevic M, Fluca AL, Corsi Romanelli MM, La Sala L, Dozio E. The Advanced Glycation End-Products (AGE)-Receptor for AGE System (RAGE): An Inflammatory Pathway Linking Obesity and Cardiovascular Diseases. Int J Mol Sci 2025; 26:3707. [PMID: 40332316 PMCID: PMC12028226 DOI: 10.3390/ijms26083707] [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: 02/25/2025] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
The AGE (advanced glycation end-products)-RAGE (receptor for AGE) system is a pro-inflammatory pathway that contributes to the pathogenesis of obesity and obesity-related cardiovascular disorders (CVD). Circulating AGE and the soluble form of RAGE (sRAGE) has been suggested as a potential biomarker of CVD related to obesity. In this study, we aim to (1) summarize the current knowledge about the role of obesity in the onset and progression of CVD, (2) discuss the role of the AGE-RAGE system as a pathway promoting obesity and linking obesity to CVD, and (3) highlight available strategies for reducing AGE-RAGE system activation and the associated beneficial effects.
Collapse
Affiliation(s)
- Elena Vianello
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- Experimental Laboratory for Research on Organ Damage Biomarkers, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Antonio P. Beltrami
- Department of Medicine, Università degli Studi di Udine, 33100 Udine, Italy;
- Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Aneta Aleksova
- Department of Medical Surgical and Health Sciences, Università degli Studi di Trieste, 34129 Trieste, Italy; (A.A.); (M.J.); (A.L.F.)
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, 34100 Trieste, Italy
| | - Milijana Janjusevic
- Department of Medical Surgical and Health Sciences, Università degli Studi di Trieste, 34129 Trieste, Italy; (A.A.); (M.J.); (A.L.F.)
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, 34100 Trieste, Italy
| | - Alessandra L. Fluca
- Department of Medical Surgical and Health Sciences, Università degli Studi di Trieste, 34129 Trieste, Italy; (A.A.); (M.J.); (A.L.F.)
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, 34100 Trieste, Italy
| | - Massimiliano M. Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- Department of Clinical and Experimental Pathology, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Lucia La Sala
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- IRCCS Multimedica, 20138 Milan, Italy
| | - Elena Dozio
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- Experimental Laboratory for Research on Organ Damage Biomarkers, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| |
Collapse
|
3
|
Mohamed AA, Al-Obeidat F, Abdallah GM, Ibrahim IT, Ali NS, Hussein MA, Hafez W, Girgiss MW, Shalby H, El-Bohy D, Elgamal R, Farghly MI, Shaheen MM, Elmahdy R, Nagaty RA, Hassan NAIF, Hamdi A, Mahmoud MO. RAGE gene polymorphism (rs1800625) and type 1 diabetes mellitus: A potential new model for early diagnosis and risk prediction. NARRA J 2025; 5:e1603. [PMID: 40352233 PMCID: PMC12059820 DOI: 10.52225/narra.v5i1.1603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/19/2025] [Indexed: 05/14/2025]
Abstract
Studies have associated advanced glycation end-products (AGEs) and the polymorphism of the AGEs receptor (RAGE) gene with clinical disorders, such as diabetes, in certain ethnic groups. However, its association with type 1 diabetes mellitus (T1DM) in Egyptians has not yet been explored. The aim of this study was to investigate the association between the RAGE gene polymorphism rs1800625 and T1DM susceptibility in Egyptians. A case- control study was conducted with 177 T1DM patients and 177 age- and sex-matched healthy controls. Variables included glycemic markers (fasting blood glucose (FBG), postprandial blood glucose (PBG), hemoglobin A1c (HbA1c)), anthropometric measurements (waist circumference, body mass index (BMI)), lipid profile (total cholesterol, triglycerides, high-density lipoprotein (HDL), low-density lipoprotein (LDL)), renal function (albumin-to-creatinine ratio (A/C ratio), serum creatinine), and history of hypertension and smoking. Genotype distribution and allele frequency of the RAGE rs1800625 polymorphism (TT, TC, CC genotypes; T and C alleles) were assessed. This study identified the RAGE rs1800625 polymorphism as a significant genetic factor associated with T1DM susceptibility. The CC genotype was significantly more prevalent in patients compared to controls (29.9% vs 11.9%; OR: 3.62; 95%CI: 1.87-6.97; p < 0.001). Similarly, the C allele was more common in patients (54.5% vs 41.0%, OR: 1.73; 95%CI: 1.28-2.33; p < 0.001). Multivariate analysis revealed that HbA1c (adjusted OR (aOR): 12.97; 95%CI: 4.00-42.05; p < 0.001), FBG (aOR: 8.96; 95%CI: 1.59-50.47; p = 0.010), and the rs1800625 polymorphism (aOR: 1.82; 95%CI: 1.146-2.876; p = 0.010) were significant predictors of T1DM. In conclusion, a genetic association was found between the RAGE gene polymorphism rs1800625 and T1DM susceptibility, with the CC genotype and C allele being more common in T1DM patients. FBG, HbA1c, and rs1800625 were identified as key predictors for T1DM, with HbA1c being the strongest. These findings highlight the importance of integrating genetic and metabolic factors in managing T1DM.
Collapse
Affiliation(s)
- Amal A. Mohamed
- Department of Biochemistry and Molecular Biology, National Hepatology and Tropical Medicine Research Institute, General Organization of Teaching Hospitals and Institutions, Cairo, Egypt
| | - Feras Al-Obeidat
- College of Technological Innovation, Zayed University, Abu Dhabi, United Arab Emirates
| | - Gamil M. Abdallah
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Ibrahim T. Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Nada S. Ali
- Department of Internal Medicine, National Institute of Diabetes and Endocrinology, General Organization of Teaching Hospitals and Institutions, Cairo, Egypt
| | - Mona A. Hussein
- Department of Internal Medicine, National Institute of Diabetes and Endocrinology, General Organization of Teaching Hospitals and Institutions, Cairo, Egypt
| | - Wael Hafez
- Medical Research and Clinical Studies Institute, The National Research Centre, Cairo, Egypt
| | - Mina W. Girgiss
- Medical Research and Clinical Studies Institute, The National Research Centre, Cairo, Egypt
| | - Hassan Shalby
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Cairo, Egypt
| | - Doaa El-Bohy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rasha Elgamal
- Department of Clinical Pathology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Maysa I. Farghly
- Department of Clinical Pathology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Mahmoud M. Shaheen
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Reem Elmahdy
- Department of Internal Medicine, Faculty of Medicine, Suez University, Suez, Egypt
| | - Raghda A. Nagaty
- Department of Clinical and Chemical Pathology, Research Institute of Ophthalmology, Giza, Egypt
| | | | - Amel Hamdi
- Hematology and Molecular Biology, Health Sciences, College of Health Science, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Mohamed O. Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
4
|
Sarkar S. Pathological role of RAGE underlying progression of various diseases: its potential as biomarker and therapeutic target. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3467-3487. [PMID: 39589529 DOI: 10.1007/s00210-024-03595-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor with several structural types, performing a myriad of molecular mechanisms. The RAGE-ligand interactions play important roles in maintaining latent chronic inflammation, and oxidative damage underlying various pathological conditions like metabolic syndrome (MetS), neurodegenerative diseases, stroke, cardiovascular disorders, pulmonary disorders, cancer and infections. RAGE is thoroughly explored in knockout animals and human trials, targeted by small molecule inhibitors, peptides, diet, and natural compounds. But it is yet to be incorporated in the mainstream management of any ailment. This review performs an appraisal of the pathological mechanisms influenced by RAGE to uncover its prospects as a biomarker while also assessing its power to become a promising therapeutic target.
Collapse
Affiliation(s)
- Sinjini Sarkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be-University, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
5
|
Ho CN, Ayers AT, Beisswenger P, Chalew S, Schmidt AM, Pandey A, Kapahi P, Fleming A, Klonoff DC. Advanced Glycation End Products (AGEs) Webinar Meeting Report. J Diabetes Sci Technol 2025; 19:576-581. [PMID: 39508279 PMCID: PMC11571634 DOI: 10.1177/19322968241296541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The advanced glycation end products (AGEs) Webinar was co-hosted by Diabetes Technology Society and Kitalys Institute on August 8, 2024, with the goal of reviewing progress made in the measurement and use of AGEs in clinical practice. Meeting topics included (1) AGEs as predictors of diabetic nephropathy (DKD), (2) hemoglobin glycation index (HGI) and the glycation gap (GG), (3) formation and structure of AGEs, (4) AGEs as a risk factor of cardiovascular disease (CVD), and (5) approaches to limit or prevent AGE formation.
Collapse
Affiliation(s)
- Cindy N. Ho
- Diabetes Technology Society, Burlingame, CA, USA
| | | | - Paul Beisswenger
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Journey Biosciences, Cleveland, OH, USA
| | - Stuart Chalew
- Section Pediatric Endocrinology/Diabetes, School of Medicine, LSU Health, The Children’s Hospital of New Orleans, New Orleans, LA, USA
| | - Ann Marie Schmidt
- Department of Medicine, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Ambarish Pandey
- Division of Cardiology and Geriatrics, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA, USA
- Juvify Bio, Novato, CA, USA
| | - Alexander Fleming
- Kinexum, Harpers Ferry, WV, USA
- Kitalys Institute, Charlottesville, VA, USA
| | - David C. Klonoff
- Diabetes Research Institute, Mills-Peninsula Medical Center, San Mateo, CA, USA
| |
Collapse
|
6
|
Li C, Liu Y, Liu C, Chen F, Xie Y, Zeh HJ, Yu C, Liu J, Tang D, Kang R. AGER-dependent macropinocytosis drives resistance to KRAS-G12D-targeted therapy in advanced pancreatic cancer. Sci Transl Med 2025; 17:eadp4986. [PMID: 39879317 DOI: 10.1126/scitranslmed.adp4986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/07/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) driven by the KRAS-G12D mutation presents a formidable health challenge because of limited treatment options. MRTX1133 is a highly selective and first-in-class KRAS-G12D inhibitor under clinical development. Here, we report that the advanced glycosylation end product-specific receptor (AGER) plays a key role in mediating MRTX1133 resistance in PDAC cells. The up-regulation of AGER within cancer cells instigates macropinocytosis, facilitating the internalization of serum albumin and subsequent amino acid generation. These amino acids are then used to synthesize the antioxidant glutathione, leading to resistance to MRTX1133 treatment due to the inhibition of apoptosis. The underlying molecular mechanism involves AGER's interaction with diaphanous-related formin 1 (DIAPH1), a formin protein responsible for driving Rac family small GTPase 1 (RAC1)-dependent macropinosome formation. The effectiveness and safety of combining MRTX1133 with pharmacological inhibitors of the AGER-DIAPH1 complex (using RAGE299) or macropinocytosis (using EIPA) were confirmed in patient-derived xenografts, orthotopic models, and genetically engineered mouse PDAC models. This combination therapy also induces high-mobility group box 1 (HMGB1) release, resulting in a subsequent antitumor CD8+ T cell response in immunocompetent mice. Collectively, the study findings underscore the potential to enhance the efficacy of KRAS-G12D blockade therapy by targeting AGER-dependent macropinocytosis.
Collapse
Affiliation(s)
- Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Fangquan Chen
- DAMP Laboratory, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Yangchun Xie
- Department of Oncology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
7
|
Menon RG, Yepuri G, Martel D, Quadri N, Hasan SN, Manigrasso MB, Shekhtman A, Schmidt AM, Ramasamy R, Regatte RR. Assessment of cardiac and skeletal muscle metabolites using 1H-MRS and chemical-shift encoded magnetic resonance imaging: Impact of diabetes, RAGE, and DIAPH1. NMR IN BIOMEDICINE 2025; 38:e5275. [PMID: 39468867 DOI: 10.1002/nbm.5275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024]
Abstract
Diabetes affects metabolism and metabolite concentrations in multiple organs. Previous preclinical studies have shown that receptor for advanced glycation end products (RAGE, gene symbol Ager) and its cytoplasmic domain binding partner, Diaphanous-1 (DIAPH1), are key mediators of diabetic micro- and macro-vascular complications. In this study, we used 1H-Magnetic Resonance Spectroscopy (MRS) and chemical shift encoded (CSE) Magnetic Resonance Imaging (MRI) to investigate the metabolite and water-fat fraction in the heart and hind limb muscle in a murine model of type 1 diabetes (T1D) and to determine if the metabolite changes in the heart and hind limb are influenced by (a) deletion of Ager or Diaph1 and (b) pharmacological blockade of RAGE-DIAPH1 interaction in mice. Nine cohorts of male mice, with six mice per cohort, were used: wild type non-diabetic control mice (WT-NDM), WT-diabetic (WT-DM) mice, Ager knockout non-diabetic (RKO-NDM) and diabetic mice (RKO-DM), Diaph1 knockout non-diabetic (DKO-NDM), and diabetic mice (DKO-DM), WT-NDM mice treated with vehicle, WT-DM mice treated with vehicle, and WT-DM mice treated with RAGE229 (antagonist of RAGE-DIAPH1 interaction). A Point Resolved Spectroscopy (PRESS) sequence for 1H-MRS, and multi-echo gradient recalled echo (GRE) for CSE were employed. Triglycerides, and free fatty acids in the heart and hind limb obtained from MRS and MRI were compared to those measured using biochemical assays. Two-sided t-test, non-parametric Kruskal-Wallis Test, and one-way ANOVA were employed for statistical analysis. We report that the results were well-correlated with significant differences using MRI and biochemical assays between WT-NDM and WT-DM, as well as within the non-diabetic groups, and within the diabetic groups. Deletion of Ager or Diaph1, or treatment with RAGE229 attenuated diabetes-associated increases in triglycerides in the heart and hind limb in mice. These results suggest that the employment of 1H-MRS/MRI is a feasible non-invasive modality to monitor metabolic dysfunction in T1D and the metabolic consequences of interventions that block RAGE and DIAPH1.
Collapse
Affiliation(s)
- Rajiv G Menon
- Department of Radiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Gautham Yepuri
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Dimitri Martel
- Department of Radiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Nosirudeen Quadri
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Syed Nurul Hasan
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Michaele B Manigrasso
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Ravinder R Regatte
- Department of Radiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
8
|
Liu J, Wang Y, Zeng L, Yu C, Kang R, Klionsky DJ, Jiang J, Tang D. Extracellular NCOA4 is a mediator of septic death by activating the AGER-NFKB pathway. Autophagy 2024; 20:2616-2631. [PMID: 38916095 PMCID: PMC11587848 DOI: 10.1080/15548627.2024.2372215] [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: 03/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024] Open
Abstract
Sepsis, a life-threatening condition resulting from a dysregulated response to pathogen infection, poses a significant challenge in clinical management. Here, we report a novel role for the autophagy receptor NCOA4 in the pathogenesis of sepsis. Activated macrophages and monocytes secrete NCOA4, which acts as a mediator of septic death in mice. Mechanistically, lipopolysaccharide, a major component of the outer membrane of Gram-negative bacteria, induces NCOA4 secretion through autophagy-dependent lysosomal exocytosis mediated by ATG5 and MCOLN1. Moreover, bacterial infection with E. coli or S. enterica leads to passive release of NCOA4 during GSDMD-mediated pyroptosis. Upon release, extracellular NCOA4 triggers the activation of the proinflammatory transcription factor NFKB/NF-κB by promoting the degradation of NFKBIA/IκB molecules. This process is dependent on the pattern recognition receptor AGER, rather than TLR4. In vivo studies employing endotoxemia and polymicrobial sepsis mouse models reveal that a monoclonal neutralizing antibody targeting NCOA4 or AGER delays animal death, protects against organ damage, and attenuates systemic inflammation. Furthermore, elevated plasma NCOA4 levels in septic patients, particularly in non-survivors, correlate positively with the sequential organ failure assessment score and concentrations of lactate and proinflammatory mediators, such as TNF, IL1B, IL6, and HMGB1. These findings demonstrate a previously unrecognized role of extracellular NCOA4 in inflammation, suggesting it as a potential therapeutic target for severe infectious diseases. Abbreviation: BMDMs: bone marrow-derived macrophages; BUN: blood urea nitrogen; CLP: cecal ligation and puncture; ELISA: enzyme-linked immunosorbent assay; LPS: lipopolysaccharide; NO: nitric oxide; SOFA: sequential organ failure assessment.
Collapse
Affiliation(s)
- Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yichun Wang
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Chongqing, China
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Chongqing, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Liu J, Li H, Chen H, Xiao X, Jin Z, Paerhati P, Bao W, Cui C, Zhu J, Yuan Y. An anti-RAGE chimeric antibody alleviates CCl 4-induced liver fibrosis via RAGE/NF-kB pathway in mice. Biomed Pharmacother 2024; 181:117737. [PMID: 39657505 DOI: 10.1016/j.biopha.2024.117737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024] Open
Abstract
Liver fibrosis is a progressive condition characterized by excessive deposition of extracellular matrix components, leading to organ dysfunction. Chronic inflammation and activation of hepatic stellate cells (HSCs) are two dominant events in all stages of fibrosis development. The receptor for advanced glycation end products (RAGE) pathway is involved in modulating liver injury and fibrosis, and preventing it, or deletion of Ager gene can protect the liver against fibrosis progression. To investigate functions and mechanism of chimeric anti-RAGE monoclonal antibody against liver fibrosis, murine-derived monoclonal anti-RAGE antibodies were used to construct murine-human chimeric antibodies. The properties of the chimeric antibody were characterized, and the biological functions of antibody A5 or its evolved humanized molecule, huA5, were investigated in cell or animal model. The data showed that blocking the RAGE pathway with huA5 robustly reduced liver injury and fibrosis. Furthermore, huA5 significantly suppressed the activation of HSCs and inhibited expression of fibrosis-associated genes, including COL1A1,TIMP1, and ACTA2. huA5 also interfered with RAGE downstream signal transduction and down-regulate both ERK and NF-κB phosphorylation, inhibited the RAGE/NF-kB pathway, leading to reduced expression of pro-inflammatory cytokines and profibrotic markers. Finally, RAGE silencing significantly decreased the expression of activation-related genes in HSCs, inhibiting HSCs proliferation and migration. These results clearly revealed that the anti-RAGE chimeric antibody exerted antifibrotic efficacy in vitro and attenuated liver fibrosis in vivo. HuA5 can be further developed as a lead molecule of drug to treat patients with liver fibrosis.
Collapse
Affiliation(s)
- Jing Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Huiyi Li
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Hui Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Xinyi Xiao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Zhedong Jin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Pameila Paerhati
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Wenxin Bao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Caixia Cui
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Yunsheng Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| |
Collapse
|
10
|
Zglejc-Waszak K, Jozwik M, Thoene M, Wojtkiewicz J. Role of Receptor for Advanced Glycation End-Products in Endometrial Cancer: A Review. Cancers (Basel) 2024; 16:3192. [PMID: 39335163 PMCID: PMC11430655 DOI: 10.3390/cancers16183192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Endometrial cancer (EC) is the most common gynecological malignancy. EC is associated with metabolic disorders that may promote non-enzymatic glycation and activate the receptor for advanced glycation end-products (RAGE) signaling pathways. Thus, we assumed that RAGE and its ligands may contribute to EC. Of particular interest is the interaction between diaphanous-related formin 1 (Diaph1) and RAGE during the progression of human cancers. Diaph1 is engaged in the proper organization of actin cytoskeletal dynamics, which is crucial in cancer invasion, metastasis, angiogenesis, and axonogenesis. However, the detailed molecular role of RAGE in EC remains uncertain. In this review, we discuss epigenetic factors that may play a key role in the RAGE-dependent endometrial pathology. We propose that DNA methylation may regulate the activity of the RAGE pathway in the uterus. The accumulation of negative external factors, such as hyperglycemia, inflammation, and oxidative stress, may interfere with the DNA methylation process. Therefore, further research should take into account the role of epigenetic mechanisms in EC progression.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Anatomy, Faculty of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Marcin Jozwik
- Department of Gynecology and Obstetrics, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-045 Olsztyn, Poland
| | - Michael Thoene
- Department of Medical Biology, Faculty of Health Sciences, University of Warmia and Mazury in Olsztyn, Żołnierska 14C Str., 10-561 Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| |
Collapse
|
11
|
Mushajiang M, Li Y, Sun Z, Liu J, Zhang L, Wang Z. USP10 alleviates Nε-carboxymethyl-lysine-induced vascular calcification and atherogenesis in diabetes mellitus by promoting AMPK activation. Cell Signal 2024; 120:111211. [PMID: 38705504 DOI: 10.1016/j.cellsig.2024.111211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/21/2024] [Accepted: 05/03/2024] [Indexed: 05/07/2024]
Abstract
Vascular calcification (VC) is a characteristic feature in patients with diabetes mellitus (DM) and is closely associated with the osteogenic differentiation of vascular smooth muscle cells (VSMCs). Ubiquitin-Specific Protease 10 (USP10) has been shown to regulate multiple cellular processes; however, its relationship with diabetic VC remains unclear. This study aims to elucidate the role of USP10 in VC development and the underlying regulatory mechanisms. Nε-carboxymethyl lysine (CML) was significantly increased in calcified ateries from diabetic atherosclerosis ApoE-/- mice fed with high-fat diets. CML downregulated USP10 expression in VSMCs and calcified mice coronary arteries, as assessd by Western blotting, RT-qPCR,immunofluorescence and immunohistochemistry. Loss-and gain-of-function experiments were conducted both in vitro and in vivo to verify the biological functions of USP10. Ectopic expression of USP10 mitigated the severity of VC. With regard to the mechanism, the interaction between USP10 and AMPKα was investigated through double-label immunofluorescence and Co-immunoprecipitation. In vitro ubiquitination assay revealed that USP10 was capable of mediating AMPKα ubiquitination and caused increased AMPKα phosphorylation level at Thr172. Moreover, the anticalcification effect of USP10 was reversed by pharmacological inhibition of AMPK signaling pathway. The current fundings suggest an important role of USP10 in diabetic VC progression, at least in part, via mediating the ubiquitination and activation of AMPKα. USP10 may serve as a novel therapeutic target for the treatment of diabetic VC.
Collapse
MESH Headings
- Animals
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin Thiolesterase/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Mice
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Lysine/metabolism
- Lysine/analogs & derivatives
- AMP-Activated Protein Kinases/metabolism
- Male
- Ubiquitination
- Mice, Inbred C57BL
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
Collapse
Affiliation(s)
- Mayibai Mushajiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yalan Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Jia Liu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| |
Collapse
|
12
|
Dong W, Yang X, Li X, Wei S, An C, Zhang J, Shi X, Dong S. Investigation of N-Glycan Functions in Receptor for Advanced Glycation End Products V Domain through Chemical Glycoprotein Synthesis. J Am Chem Soc 2024; 146:18270-18280. [PMID: 38917169 DOI: 10.1021/jacs.4c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The receptor for advanced glycation end products (RAGE) plays a crucial role in inflammation-related pathways and various chronic diseases. Despite the recognized significance of N-glycosylation in the ligand-binding V domain (VD) of RAGE, a comprehensive understanding of the site-activity and structure-activity relationships is lacking due to the challenges in obtaining homogeneous glycoprotein samples through biological expression. Here, we combined chemical and chemoenzymatic approaches to synthesize RAGE-VD and its congeners with Asn3-glycosylation by incorporating precise N-glycan structures. Evaluation of these samples revealed that, in comparison to other RAGE-VD forms, α2,6-sialylated N-glycosylation at the Asn3 site results in more potent inhibition of HMGB1-induced nuclear factor-κB (NF-κB) expression in RAGE-overexpressing cells. Hydrogen/deuterium exchange-mass spectrum analysis revealed a sialylated RAGE-VD-induced interaction region within HMGB1. Conversely, Asn3 N-glycosylation in VD has negligible effects on RAGE-VD/S100B interactions. This study established an approach for accessing homogeneously glycosylated RAGE-VD and explored the modulatory effects of N-glycosylation on the interactions between RAGE-VD and its ligand proteins.
Collapse
Affiliation(s)
- Weidong Dong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xingyue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinyu Li
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Sheng Wei
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chuanjing An
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jun Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaomeng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Suwei Dong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and Department of Chemical Biology at School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
13
|
Ramasamy R, Shekhtman A, Schmidt AM. RAGE/DIAPH1 Axis and Cardiometabolic Disease: From Nascent Discoveries to Therapeutic Potential. Arterioscler Thromb Vasc Biol 2024; 44:1497-1501. [PMID: 38924438 PMCID: PMC11210684 DOI: 10.1161/atvbaha.124.320142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Medical Center (R.R., A.M.S.)
| | | | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Medical Center (R.R., A.M.S.)
| |
Collapse
|
14
|
Han Z, Li A, Xue Z, Guan SB, Yin G, Zheng X. Eugenol-loaded polyurethane gelatin dressing for efficient angiogenesis and antibacterial effects in refractory diabetic wound defect healing. Int J Biol Macromol 2024; 271:132619. [PMID: 38795896 DOI: 10.1016/j.ijbiomac.2024.132619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
The amelioration of refractory diabetic ulcers presents a formidable conundrum on a global scale, attributable to the elevated peril of contagion and protracted convalescence durations. Within the purlieus of this reparative epoch, the deployment of efficacious wound coverings endowed with both angiogenesis and antibacterial attributes is of paramount significance. Hydrogel wound dressings are distinguished by their elevated biocompatibility, adhesive tenacity, and innate regenerative capacity. Eugenol, a substance distilled from the blossoms of the lilac, serves as a precursor to metformin and is known to impede the genesis of reactive oxygen species. Although its antibacterial effects have been extensively chronicled, the angiogenic ramifications of eugenol within the context of wound remediation remain under-investigated. This research aimed to evaluate the effectiveness of eugenol-infused hydrogel as a wound dressing material. In this context, polyurethane gelatin (PG) was combined with eugenol at concentrations of 0.5% and 1%, creating PG-eugenol hydrogel mixtures with specific mass ratios for both in vivo and in vitro assessments. The in vivo studies indicated that hydrogels infused with eugenol expedited diabetic wound healing by fostering angiogenesis. Enhanced healing was noted, attributed to improved antibacterial and angiogenic properties, increased cell proliferation, tissue regeneration, and re-epithelialization. The in vitro analyses revealed that eugenol-enriched hydrogels stimulated the growth of fibroblasts (HFF-1) and human umbilical vein endothelial cells (HUVECs) and exhibited antibacterial characteristics. This investigation confirms the potential of eugenol-laden hydrogels in effectively treating diabetic wound defects.
Collapse
Affiliation(s)
- Zhengzhe Han
- National Center for Orthopaedics, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ang Li
- National Center for Orthopaedics, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zichao Xue
- Department of Sports Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Shi-Bing Guan
- Department of Hand and Foot Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China.
| | - Gang Yin
- Trauma Center, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| | - Xianyou Zheng
- National Center for Orthopaedics, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
15
|
Fei X, Wang L, Dou YN, Fei F, Zhang Y, Lv W, He X, Wu X, Chao W, Chen H, Wei J, Gao D, Fei Z. Extracellular vesicle encapsulated Homer1a as novel nanotherapeutics against intracerebral hemorrhage in a mouse model. J Neuroinflammation 2024; 21:85. [PMID: 38582897 PMCID: PMC10999083 DOI: 10.1186/s12974-024-03088-6] [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: 10/14/2023] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
Homer1a and A2 astrocytes are involved in the regulation of inflammation induced by intracerebral hemorrhage (ICH). However, there is no anticipated treatment strategy based on the anti-inflammatory effect of Homer1a and A2 astrocytes. Here, we successfully induced A2 astrocytes in vitro, and then we report an efficient method to prepare Homer1a+ EVs derived from A2 astrocytes which making it more stable, safe, and targetable to injured neurons. Homer1a+ EVs promotes the conversion of A1 to A2 astrocytes in ICH mice. Homer1a+ EVs inhibits activation and nuclear translocation of NF-κB, thereby regulating transcription of IL-17A in neurons. Homer1a+ EVs inhibits the RAGE/NF-κB/IL-17 signaling pathway and the binding ability of IL-17A: IL17-AR and RAGE: DIAPH1. In addition, Homer1a+ EVs ameliorates the pathology, behavior, and survival rate in GFAPCreHomer1fl/-Homer1a± and NestinCreRAGEfl/fl ICH mice. Our study provides a novel insight and potential for the clinical translation of Homer1a+ EVs in the treatment of ICH.
Collapse
Affiliation(s)
- Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Li Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Ya-Nan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Weihao Lv
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Wangshu Chao
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| | - Dakuan Gao
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| |
Collapse
|
16
|
Nowicka N, Zglejc-Waszak K, Juranek J, Korytko A, Wąsowicz K, Chmielewska-Krzesińska M, Wojtkiewicz J. Novel insights into RAGE signaling pathways during the progression of amyotrophic lateral sclerosis in RAGE-deficient SOD1 G93A mice. PLoS One 2024; 19:e0299567. [PMID: 38457412 PMCID: PMC10923448 DOI: 10.1371/journal.pone.0299567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is neurodegenerative disease characterized by a progressive loss of motor neurons resulting in paralysis and muscle atrophy. One of the most prospective hypothesis on the ALS pathogenesis suggests that excessive inflammation and advanced glycation end-products (AGEs) accumulation play a crucial role in the development of ALS in patients and SOD1 G93A mice. Hence, we may speculate that RAGE, receptor for advanced glycation end-products and its proinflammatory ligands such as: HMGB1, S100B and CML contribute to ALS pathogenesis. The aim of our studies was to decipher the role of RAGE as well as provide insight into RAGE signaling pathways during the progression of ALS in SOD1 G93A and RAGE-deficient SOD1 G93A mice. In our study, we observed alternations in molecular pattern of proinflammatory RAGE ligands during progression of disease in RAGE KO SOD1 G93A mice compared to SOD1 G93A mice. Moreover, we observed that the amount of beta actin (ACTB) as well as Glial fibrillary acidic protein (GFAP) was elevated in SOD1 G93A mice when compared to mice with deletion of RAGE. These data contributes to our understanding of implications of RAGE and its ligands in pathogenesis of ALS and highlight potential targeted therapeutic interventions at the early stage of this devastating disease. Moreover, inhibition of the molecular cross-talk between RAGE and its proinflammatory ligands may abolish neuroinflammation, gliosis and motor neuron damage in SOD1 G93A mice. Hence, we hypothesize that attenuated interaction of RAGE with its proinflammatory ligands may improve well-being and health status during ALS in SOD1 G93A mice. Therefore, we emphasize that the inhibition of RAGE signaling pathway may be a therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Natalia Nowicka
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Judyta Juranek
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Krzysztof Wąsowicz
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Małgorzata Chmielewska-Krzesińska
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
17
|
Guan H, Tian J, Wang Y, Niu P, Zhang Y, Zhang Y, Fang X, Miao R, Yin R, Tong X. Advances in secondary prevention mechanisms of macrovascular complications in type 2 diabetes mellitus patients: a comprehensive review. Eur J Med Res 2024; 29:152. [PMID: 38438934 PMCID: PMC10910816 DOI: 10.1186/s40001-024-01739-1] [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: 12/04/2023] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) poses a significant global health burden. This is particularly due to its macrovascular complications, such as coronary artery disease, peripheral vascular disease, and cerebrovascular disease, which have emerged as leading contributors to morbidity and mortality. This review comprehensively explores the pathophysiological mechanisms underlying these complications, protective strategies, and both existing and emerging secondary preventive measures. Furthermore, we delve into the applications of experimental models and methodologies in foundational research while also highlighting current research limitations and future directions. Specifically, we focus on the literature published post-2020 concerning the secondary prevention of macrovascular complications in patients with T2DM by conducting a targeted review of studies supported by robust evidence to offer a holistic perspective.
Collapse
Affiliation(s)
- Huifang Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Ying Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ping Niu
- Rehabilitation Department, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Ruiyang Yin
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
18
|
Arivazhagan L, Popp CJ, Ruiz HH, Wilson RA, Manigrasso MB, Shekhtman A, Ramasamy R, Sevick MA, Schmidt AM. The RAGE/DIAPH1 axis: mediator of obesity and proposed biomarker of human cardiometabolic disease. Cardiovasc Res 2024; 119:2813-2824. [PMID: 36448548 PMCID: PMC11484493 DOI: 10.1093/cvr/cvac175] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/07/2023] Open
Abstract
Overweight and obesity are leading causes of cardiometabolic dysfunction. Despite extensive investigation, the mechanisms mediating the increase in these conditions are yet to be fully understood. Beyond the endogenous formation of advanced glycation endproducts (AGEs) in overweight and obesity, exogenous sources of AGEs accrue through the heating, production, and consumption of highly processed foods. Evidence from cellular and mouse model systems indicates that the interaction of AGEs with their central cell surface receptor for AGE (RAGE) in adipocytes suppresses energy expenditure and that AGE/RAGE contributes to increased adipose inflammation and processes linked to insulin resistance. In human subjects, the circulating soluble forms of RAGE, which are mutable, may serve as biomarkers of obesity and weight loss. Antagonists of RAGE signalling, through blockade of the interaction of the RAGE cytoplasmic domain with the formin, Diaphanous-1 (DIAPH1), target aberrant RAGE activities in metabolic tissues. This review focuses on the potential roles for AGEs and other RAGE ligands and RAGE/DIAPH1 in the pathogenesis of overweight and obesity and their metabolic consequences.
Collapse
Affiliation(s)
- Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, Science Building, 435 E. 30th Street, New York, NY 10016, USA
| | - Collin J Popp
- Center for Healthful Behavior Change, Department of Population Health, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Henry H Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, Science Building, 435 E. 30th Street, New York, NY 10016, USA
| | - Robin A Wilson
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, Science Building, 435 E. 30th Street, New York, NY 10016, USA
| | - Michaele B Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, Science Building, 435 E. 30th Street, New York, NY 10016, USA
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY 12222, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, Science Building, 435 E. 30th Street, New York, NY 10016, USA
| | - Mary Ann Sevick
- Center for Healthful Behavior Change, Department of Population Health, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, Science Building, 435 E. 30th Street, New York, NY 10016, USA
| |
Collapse
|
19
|
Cai Z, Li Y, Bai L, Xu J, Liu Z, Zhang T, Gao S, Lin Y. Tetrahedral Framework Nucleic Acids Based Small Interfering RNA Targeting Receptor for Advanced Glycation End Products for Diabetic Complications Treatment. ACS NANO 2023; 17:22668-22683. [PMID: 37751401 DOI: 10.1021/acsnano.3c06999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Complications arising from diabetes can threaten multiple organs. Advanced glycation end products (AGEs) play a significant role in inducing these complications. Highly processed diets and hyperglycemia facilitate the accumulation of AGEs in the body. Interaction between AGEs and their main receptor (RAGE) initiates the transmission of intracellular inflammatory and cell death signals, which ultimately lead to complications. To counter AGEs-induced damage, we developed an siRNA-binding tetrahedral framework nucleic acids (TDN) system, termed Tsi, which combines the potent cell membrane penetrability and serum stability of TDN with the gene-targeting specificity of siRNA-RAGE. Tsi effectively and persistently downregulates the expression of RAGE, thereby suppressing inflammation by blocking the NF-κB pathway as well as exhibiting antioxidant functions. Furthermore, Tsi regulates the pyroptosis state of macrophages via the NLRP3/caspase-1 axis, which inhibits the spread of cell death signals and maintains homeostasis. This is of great significance for the synergistic treatment strategy for systemic complications in patients with refractory hyperglycemia. In summary, this study describes a nanomedicine that targets the RAGE and suppresses AGE-induced inflammation. This nucleic acid drug holds long-lasting efficacy and is independent of lowering hyperglycemia, which provides a strategy for the treatment of diabetic complications and age-related diseases.
Collapse
Affiliation(s)
- Zhengwen Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yong Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Long Bai
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jiangshan Xu
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shaojingya Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| |
Collapse
|
20
|
Carr KD, Weiner SP, Vasquez C, Schmidt AM. Involvement of the Receptor for Advanced Glycation End Products (RAGE) in high fat-high sugar diet-induced anhedonia in rats. Physiol Behav 2023; 271:114337. [PMID: 37625475 PMCID: PMC10592025 DOI: 10.1016/j.physbeh.2023.114337] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Clinical and basic science investigation indicates a link between insulin resistance and anhedonia. Previous results of this laboratory point to impaired nucleus accumbens (NAc) insulin signaling as an underpinning of diet-induced anhedonia, based on use of a glucose lick microstructure assay. The present study evaluated whether advanced glycation end products (AGEs) and their receptor (RAGE), known to mediate obesogenic diet-induced inflammation and pathological metabolic conditions, are involved in this behavioral change. Six weeks maintenance of male and female rats on a high fat-high sugar liquid diet (chocolate Ensure) increased body weight gain, and markedly increased circulating insulin and leptin, but induced anhedonia (decreased first minute lick rate and lick burst size) in males only. In these subjects, anhedonia correlated with plasma concentrations of insulin. Although the diet did not alter plasma or NAc AGEs, or the expression of RAGE in the NAc, marginally significant correlations were seen between anhedonia and plasma content of several AGEs and NAc RAGE. Importantly, a small molecule RAGE antagonist, RAGE229, administered twice daily by oral gavage, prevented diet-induced anhedonia. This beneficial effect was associated with improved adipose function, reflected in the adiponectin/leptin ratio, and increased pCREB/total CREB in the NAc, and a shift in the pCREB correlation with pThr34-DARPP-32 from near-zero to strongly positive, such that both phospho-proteins correlated with the rescued hedonic response. This set of findings suggests that the receptor/signaling pathway and cell type underlying the RAGE229-mediated increase in pCREB may mediate anhedonia and its prevention. The possible role of adipose tissue as a locus of diet-induced RAGE signaling, and source of circulating factors that target NAc to modify hedonic reactivity are discussed.
Collapse
Affiliation(s)
- Kenneth D Carr
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States; Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States.
| | - Sydney P Weiner
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Carolina Vasquez
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States; Departments of Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Ann Marie Schmidt
- Departments of Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| |
Collapse
|
21
|
Perkins RK, Miranda ER, Varshney P, Farabi SS, Quinn LT, Haus JM. Effects of acute aerobic exercise on circulating sTLR and sRAGE profiles in normal- and abnormal-glucose-tolerant individuals. Physiol Rep 2023; 11:e15859. [PMID: 37985201 PMCID: PMC10659941 DOI: 10.14814/phy2.15859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/11/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023] Open
Abstract
BMI-matched normal- (NGT, n = 10, 41 ± 4y, 35.6 ± 3.0 kg/m2 ) and abnormal-glucose-tolerant (AGT, n = 16, 51 ± 3y, 34.3 ± 1.5 kg/m2 ) participants were evaluated for body composition, metabolic health (oral glucose tolerance test [OGTT]), and VO2 max. Participants also completed a treadmill walking test at 65% VO2 max for 30 min. Total sRAGE, esRAGE, sTLR2, and sTLR4 were assessed via ELISA, and cRAGE was calculated. AGT exhibited greater (p < 0.05) body fat % (+24%), fasting plasma glucose (+37%), OGTT AUC (+59%), and HOMA-IR (+55%) and lower (p < 0.05) VO2 max (-24%). sTLR2 was 33% lower in AGT than NGT (main effect, p = 0.034). However, sTLR2 did not change (p > 0.05) following AE. sTLR4 tended to be 36% lower in AGT than NGT (main effect, p = 0.096) and did not change following AE (p > 0.05). Total sRAGE and isoforms were similar (p > 0.05) between groups and did not change following AE (p > 0.05). sTLR2 was correlated with (p < 0.05) basal BG (r = -0.505) and OGTT AUC (r = -0.687). sTLR4 was correlated with basal BG (p < 0.10, r = -0.374) and OGTT AUC (p < 0.05, r = -0.402). Linear regressions were predictive of sTLRs in the basal state (sTLR2: R2 = 0.641, p = 0.01; sTLR4: R2 = 0.566, p = 0.037) and after acute exercise state (sTLR2: R2 = 0.681, p = 0.004, sTLR4: R2 = 0.568, p = 0.036).These findings show circulating sTLR profiles are disrupted in AGT and acute AE minimally modulates their levels.
Collapse
Affiliation(s)
- Ryan K. Perkins
- Department of KinesiologyCalifornia State University, ChicoChicoCaliforniaUSA
| | | | | | - Sarah S. Farabi
- Center for Human NutritionWashington University School of MedicineSt. LouisMissouriUSA
- Goldfarb School of Nursing at Barnes‐Jewish CollegeSt. LouisMissouriUSA
| | - Lauretta T. Quinn
- Department of Behavioral Health SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Jacob M. Haus
- School of KinesiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
22
|
Yepuri G, Ramirez LM, Theophall GG, Reverdatto SV, Quadri N, Hasan SN, Bu L, Thiagarajan D, Wilson R, Díez RL, Gugger PF, Mangar K, Narula N, Katz SD, Zhou B, Li H, Stotland AB, Gottlieb RA, Schmidt AM, Shekhtman A, Ramasamy R. DIAPH1-MFN2 interaction regulates mitochondria-SR/ER contact and modulates ischemic/hypoxic stress. Nat Commun 2023; 14:6900. [PMID: 37903764 PMCID: PMC10616211 DOI: 10.1038/s41467-023-42521-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/13/2023] [Indexed: 11/01/2023] Open
Abstract
Inter-organelle contact and communication between mitochondria and sarco/endoplasmic reticulum (SR/ER) maintain cellular homeostasis and are profoundly disturbed during tissue ischemia. We tested the hypothesis that the formin Diaphanous-1 (DIAPH1), which regulates actin dynamics, signal transduction and metabolic functions, contributes to these processes. We demonstrate that DIAPH1 interacts directly with Mitofusin-2 (MFN2) to shorten mitochondria-SR/ER distance, thereby enhancing mitochondria-ER contact in cells including cardiomyocytes, endothelial cells and macrophages. Solution structure studies affirm the interaction between the Diaphanous Inhibitory Domain and the cytosolic GTPase domain of MFN2. In male rodent and human cardiomyocytes, DIAPH1-MFN2 interaction regulates mitochondrial turnover, mitophagy, and oxidative stress. Introduction of synthetic linker construct, which shorten the mitochondria-SR/ER distance, mitigated the molecular and functional benefits of DIAPH1 silencing in ischemia. This work establishes fundamental roles for DIAPH1-MFN2 interaction in the regulation of mitochondria-SR/ER contact networks. We propose that targeting pathways that regulate DIAPH1-MFN2 interactions may facilitate recovery from tissue ischemia.
Collapse
Affiliation(s)
- Gautham Yepuri
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Lisa M Ramirez
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Gregory G Theophall
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Sergei V Reverdatto
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Nosirudeen Quadri
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Syed Nurul Hasan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Lei Bu
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Devi Thiagarajan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Robin Wilson
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Raquel López Díez
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Paul F Gugger
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Kaamashri Mangar
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Navneet Narula
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Stuart D Katz
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Boyan Zhou
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Huilin Li
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Aleksandr B Stotland
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Roberta A Gottlieb
- Department of Biomedical Sciences, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Alexander Shekhtman
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA.
| |
Collapse
|
23
|
Ramasamy R, Shekhtman A, Schmidt AM. RAGE/DIAPH1 and atherosclerosis through an evolving lens: Viewing the cell from the "Inside - Out". Atherosclerosis 2023; 394:117304. [PMID: 39492058 PMCID: PMC11309734 DOI: 10.1016/j.atherosclerosis.2023.117304] [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: 06/09/2023] [Revised: 08/11/2023] [Accepted: 09/15/2023] [Indexed: 08/13/2024]
Abstract
BACKGROUND AND AIMS In hyperglycemia, inflammation, oxidative stress and aging, Damage Associated Molecular Patterns (DAMPs) accumulate in conditions such as atherosclerosis. Binding of DAMPs to receptors such as the receptor for advanced glycation end products (RAGE) activates signal transduction cascades that contribute to cellular stress. The cytoplasmic domain (tail) of RAGE (ctRAGE) binds to the formin Diaphanous1 (DIAPH1), which is important for RAGE signaling. This Review will detail the evidence linking the RAGE/DIAPH1 signaling pathway to atherosclerosis and envisages future therapeutic opportunities from the "inside-out" point of view in affected cells. METHODS PubMed was searched using a variety of search terms, including "receptor for advanced glycation end products" along with various combinations including "and atherosclerosis," "soluble RAGE and atherosclerosis," "statins and RAGE," "PPAR and RAGE" and "SGLT2 inhibitor and RAGE." RESULTS In non-diabetic and diabetic mice, antagonism or global deletion of Ager (the gene encoding RAGE) retards progression and accelerates regression of atherosclerosis. Global deletion of Diaph1 in mice devoid of the low density lipoprotein receptor (Ldlr) significantly attenuates atherosclerosis; mice devoid of both Diaph1 and Ldlr display significantly lower plasma and liver concentrations of cholesterol and triglyceride compared to mice devoid of Ldlr. Associations between RAGE pathway and human atherosclerosis have been identified based on relationships between plasma/serum concentrations of RAGE ligands, soluble RAGEs and atherosclerosis. CONCLUSIONS Efforts to target RAGE/DIAPH1 signaling through a small molecule antagonist therapeutic strategy hold promise to quell accelerated atherosclerosis in diabetes and in other forms of cardiovascular disease.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Medical Center, NY, USA
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Medical Center, NY, USA.
| |
Collapse
|
24
|
Wilson RA, Arivazhagan L, Ruiz HH, Zhou B, Qian K, Manigrasso MB, Bernadin R, Mangar K, Shekhtman A, Li H, Ramasamy R, Schmidt AM. Pharmacological antagonism of receptor for advanced glycation end products signaling promotes thermogenesis, healthful body mass and composition, and metabolism in mice. Obesity (Silver Spring) 2023; 31:1825-1843. [PMID: 37231626 PMCID: PMC10790363 DOI: 10.1002/oby.23774] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Optimal body mass and composition as well as metabolic fitness require tightly regulated and interconnected mechanisms across tissues. Disturbances in these regulatory networks tip the balance between metabolic health versus overweight and obesity and their complications. The authors previously demonstrated roles for the receptor for advanced glycation end products (RAGE) in obesity, as global- or adipocyte-specific deletion of Ager (the gene encoding RAGE) protected mice from high-fat diet-induced obesity and metabolic dysfunction. METHODS To explore translational strategies evoked by these observations, a small molecule antagonist of RAGE signaling, RAGE229, was administered to lean mice and mice with obesity undergoing diet-induced weight loss. Body mass and composition and whole body and adipose tissue metabolism were examined. RESULTS This study demonstrates that antagonism of RAGE signaling reduced body mass and adiposity and improved glucose, insulin, and lipid metabolism in lean male and female mice and in male mice with obesity undergoing weight loss. In adipose tissue and in human and mouse adipocytes, RAGE229 enhanced phosphorylation of protein kinase A substrates, which augmented lipolysis, mitochondrial function, and thermogenic programs. CONCLUSIONS Pharmacological antagonism of RAGE signaling is a potent strategy to optimize healthful body mass and composition and metabolic fitness.
Collapse
Affiliation(s)
- Robin A. Wilson
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Henry H. Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Boyan Zhou
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Kun Qian
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Michaele B. Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Rollanda Bernadin
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Kaamashri Mangar
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York, Albany, New York, USA
| | - Huilin Li
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
25
|
Jiang L, Wu X, Wang Y, Liu C, Wu Y, Wang J, Xu N, He Z, Wang S, Zhang H, Wang X, Lu X, Tan Q, Sun X. Photothermal Controlled-Release Immunomodulatory Nanoplatform for Restoring Nerve Structure and Mechanical Nociception in Infectious Diabetic Ulcers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300339. [PMID: 37148168 PMCID: PMC10369251 DOI: 10.1002/advs.202300339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/04/2023] [Indexed: 05/08/2023]
Abstract
Infectious diabetic ulcers (IDU) require anti-infection, angiogenesis, and nerve regeneration therapy; however, the latter has received comparatively less research attention than the former two. In particular, there have been few reports on the recovery of mechanical nociception. In this study, a photothermal controlled-release immunomodulatory hydrogel nanoplatform is tailored for the treatment of IDU. Due to a thermal-sensitive interaction between polydopamine-reduced graphene oxide (pGO) and the antibiotic mupirocin, excellent antibacterial efficacy is achieved through customized release kinetics. In addition, Trem2+ macrophages recruited by pGO regulate collagen remodeling and restore skin adnexal structures to alter the fate of scar formation, promote angiogenesis, accompanied by the regeneration of neural networks, which ensures the recovery of mechanical nociception and may prevent the recurrence of IDU at the source. In all, a full-stage strategy from antibacterial, immune regulation, angiogenesis, and neurogenesis to the recovery of mechanical nociception, an indispensable neural function of skin, is introduced to IDU treatment, which opens up an effective and comprehensive therapy for refractory IDU.
Collapse
Affiliation(s)
- Le Jiang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Xiangyi Wu
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Yifan Wang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Chunlin Liu
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Yixian Wu
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Jingyun Wang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Nan Xu
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Zhijun He
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Shuqin Wang
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Hao Zhang
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Xiong Lu
- Key Lab of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Qian Tan
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| |
Collapse
|
26
|
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
|
27
|
Liu J, Jin Z, Wang X, Jakoš T, Zhu J, Yuan Y. RAGE pathways play an important role in regulation of organ fibrosis. Life Sci 2023; 323:121713. [PMID: 37088412 DOI: 10.1016/j.lfs.2023.121713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/09/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
Organ fibrosis is a pathological process of fibroblast activation and excessive deposition of extracellular matrix after persistent tissue injury and therefore is a common endpoint of many organ pathologies. Multiple cellular types and soluble mediators, including chemokines, cytokines and non-peptidic factors, are implicated in fibrogenesis and the remodeling of tissue architecture. The molecular basis of the fibrotic process is complex and consists of closely intertwined signaling networks. Research has strived for a better understanding of these pathological mechanisms to potentially reveal novel therapeutic targets for fibrotic diseases. In light of new knowledge, the receptor for advanced glycation end products (RAGE) emerged as an important candidate for the regulation of a wide variety of cellular functions related to fibrosis, including inflammation, cell proliferation, apoptosis, and angiogenesis. RAGE is a pattern recognition receptor that binds a broad range of ligands such as advanced glycation end products, high mobility group box-1, S-100 calcium-binding protein and amyloid beta protein. Although the link between RAGE and fibrosis has been established, the exact mechanisms need be investigated in further studies. The aim of this review is to collect all available information about the intricate function of RAGE and its signaling cascades in the pathogenesis of fibrotic diseases within different organs. In addition, to the major ligands and signaling pathways, we discuss potential strategies for targeting RAGE in fibrosis. We emphasize the functional links between RAGE, inflammation and fibrosis that may guide further studies and the development of improved therapeutic drugs.
Collapse
Affiliation(s)
- Jing Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Zhedong Jin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Xiaolong Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Tanja Jakoš
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Yunsheng Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| |
Collapse
|
28
|
Reddy VP, Aryal P, Soni P. RAGE Inhibitors in Neurodegenerative Diseases. Biomedicines 2023; 11:biomedicines11041131. [PMID: 37189749 DOI: 10.3390/biomedicines11041131] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Nonenzymatic reactions of reducing sugars with primary amino groups of amino acids, proteins, and nucleic acids, followed by oxidative degradations would lead to the formation of advanced glycation endproducts (AGEs). The AGEs exert multifactorial effects on cell damage leading to the onset of neurological disorders. The interaction of AGEs with the receptors for advanced glycation endproducts (RAGE) contribute to the activation of intracellular signaling and the expression of the pro-inflammatory transcription factors and various inflammatory cytokines. This inflammatory signaling cascade is associated with various neurological diseases, including Alzheimer's disease (AD), secondary effects of traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), and diabetic neuropathy, and other AGE-related diseases, including diabetes and atherosclerosis. Furthermore, the imbalance of gut microbiota and intestinal inflammation are also associated with endothelial dysfunction, disrupted blood-brain barrier (BBB) and thereby the onset and progression of AD and other neurological diseases. AGEs and RAGE play an important role in altering the gut microbiota composition and thereby increase the gut permeability and affect the modulation of the immune-related cytokines. The inhibition of the AGE-RAGE interactions, through small molecule-based therapeutics, prevents the inflammatory cascade of events associated with AGE-RAGE interactions, and thereby attenuates the disease progression. Some of the RAGE antagonists, such as Azeliragon, are currently in clinical development for treating neurological diseases, including AD, although currently there have been no FDA-approved therapeutics based on the RAGE antagonists. This review outlines the AGE-RAGE interactions as a leading cause of the onset of neurological diseases and the current efforts on developing therapeutics for neurological diseases based on the RAGE antagonists.
Collapse
Affiliation(s)
- V Prakash Reddy
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Puspa Aryal
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Pallavi Soni
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|
29
|
Senatus L, Egaña-Gorroño L, López-Díez R, Bergaya S, Aranda JF, Amengual J, Arivazhagan L, Manigrasso MB, Yepuri G, Nimma R, Mangar KN, Bernadin R, Zhou B, Gugger PF, Li H, Friedman RA, Theise ND, Shekhtman A, Fisher EA, Ramasamy R, Schmidt AM. DIAPH1 mediates progression of atherosclerosis and regulates hepatic lipid metabolism in mice. Commun Biol 2023; 6:280. [PMID: 36932214 PMCID: PMC10023694 DOI: 10.1038/s42003-023-04643-2] [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: 01/14/2022] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
Atherosclerosis evolves through dysregulated lipid metabolism interwoven with exaggerated inflammation. Previous work implicating the receptor for advanced glycation end products (RAGE) in atherosclerosis prompted us to explore if Diaphanous 1 (DIAPH1), which binds to the RAGE cytoplasmic domain and is important for RAGE signaling, contributes to these processes. We intercrossed atherosclerosis-prone Ldlr-/- mice with mice devoid of Diaph1 and fed them Western diet for 16 weeks. Compared to male Ldlr-/- mice, male Ldlr-/- Diaph1-/- mice displayed significantly less atherosclerosis, in parallel with lower plasma concentrations of cholesterol and triglycerides. Female Ldlr-/- Diaph1-/- mice displayed significantly less atherosclerosis compared to Ldlr-/- mice and demonstrated lower plasma concentrations of cholesterol, but not plasma triglycerides. Deletion of Diaph1 attenuated expression of genes regulating hepatic lipid metabolism, Acaca, Acacb, Gpat2, Lpin1, Lpin2 and Fasn, without effect on mRNA expression of upstream transcription factors Srebf1, Srebf2 or Mxlipl in male mice. We traced DIAPH1-dependent mechanisms to nuclear translocation of SREBP1 in a manner independent of carbohydrate- or insulin-regulated cues but, at least in part, through the actin cytoskeleton. This work unveils new regulators of atherosclerosis and lipid metabolism through DIAPH1.
Collapse
Affiliation(s)
- Laura Senatus
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Lander Egaña-Gorroño
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Sonia Bergaya
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Juan Francisco Aranda
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Jaume Amengual
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Michaele B Manigrasso
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Gautham Yepuri
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Ramesh Nimma
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Kaamashri N Mangar
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Rollanda Bernadin
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Boyan Zhou
- Department of Population Health, Division of Biostatistics, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Paul F Gugger
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Huilin Li
- Department of Population Health, Division of Biostatistics, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center and Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil D Theise
- Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, New York, USA
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY, USA
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
30
|
Clark JM, Garvey WT, Niswender KD, Schmidt AM, Ahima RS, Aleman JO, Battarbee AN, Beckman J, Bennett WL, Brown NJ, Chandler‐Laney P, Cox N, Goldberg IJ, Habegger KM, Harper LM, Hasty AH, Hidalgo BA, Kim SF, Locher JL, Luther JM, Maruthur NM, Miller ER, Sevick MA, Wells Q. Obesity and Overweight: Probing Causes, Consequences, and Novel Therapeutic Approaches Through the American Heart Association's Strategically Focused Research Network. J Am Heart Assoc 2023; 12:e027693. [PMID: 36752232 PMCID: PMC10111504 DOI: 10.1161/jaha.122.027693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/03/2023] [Indexed: 02/09/2023]
Abstract
As the worldwide prevalence of overweight and obesity continues to rise, so too does the urgency to fully understand mediating mechanisms, to discover new targets for safe and effective therapeutic intervention, and to identify biomarkers to track obesity and the success of weight loss interventions. In 2016, the American Heart Association sought applications for a Strategically Focused Research Network (SFRN) on Obesity. In 2017, 4 centers were named, including Johns Hopkins University School of Medicine, New York University Grossman School of Medicine, University of Alabama at Birmingham, and Vanderbilt University Medical Center. These 4 centers were convened to study mechanisms and therapeutic targets in obesity, to train a talented cadre of American Heart Association SFRN-designated fellows, and to initiate and sustain effective and enduring collaborations within the individual centers and throughout the SFRN networks. This review summarizes the central themes, major findings, successful training of highly motivated and productive fellows, and the innovative collaborations and studies forged through this SFRN on Obesity. Leveraging expertise in in vitro and cellular model assays, animal models, and humans, the work of these 4 centers has made a significant impact in the field of obesity, opening doors to important discoveries, and the identification of a future generation of obesity-focused investigators and next-step clinical trials. The creation of the SFRN on Obesity for these 4 centers is but the beginning of innovative science and, importantly, the birth of new collaborations and research partnerships to propel the field forward.
Collapse
Affiliation(s)
- Jeanne M. Clark
- Division of General Internal Medicine, Department of MedicineThe Johns Hopkins University School of MedicineBaltimoreMD
- Department of EpidemiologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchThe Johns Hopkins UniversityBaltimoreMD
| | - W. Timothy Garvey
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamAL
| | - Kevin D. Niswender
- Tennessee Valley Healthcare SystemVanderbilt University Medical CenterNashvilleTN
- Division of Diabetes, Department of Medicine, Endocrinology and MetabolismVanderbilt University Medical CenterNashvilleTN
| | - Ann Marie Schmidt
- Department of Medicine, Diabetes Research Program, Division of Endocrinology, Diabetes and MetabolismNew York University Grossman School of MedicineNew YorkNY
| | - Rexford S. Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and MetabolismThe Johns Hopkins University School of MedicineBaltimoreMD
| | - Jose O. Aleman
- Division of Endocrinology, Department of Medicine, Diabetes and MetabolismNew York University Grossman School of MedicineNew YorkNY
| | - Ashley N. Battarbee
- Division of Maternal Fetal Medicine, Department of Obstetrics and GynecologyUniversity of Alabama at BirminghamBirminghamAL
| | - Joshua Beckman
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTN
| | - Wendy L. Bennett
- Division of General Internal Medicine, Department of MedicineThe Johns Hopkins University School of MedicineBaltimoreMD
- Department of EpidemiologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchThe Johns Hopkins UniversityBaltimoreMD
- Department of Population, Family and Reproductive HealthThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | | | | | - Nancy Cox
- Vanderbilt Genetics Institute and Division of Genetic Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Ira J. Goldberg
- Division of Endocrinology, Department of Medicine, Diabetes and MetabolismNew York University Grossman School of MedicineNew YorkNY
| | - Kirk M. Habegger
- Division of Endocrinology, Department of Medicine, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAL
| | - Lorie M. Harper
- Division of Maternal Fetal Medicine, Department of Obstetrics and GynecologyUniversity of Alabama at BirminghamBirminghamAL
- Division of Maternal‐Fetal Medicine, Department of Women’s Health, Dell Medical SchoolUniversity of Texas at AustinAustinTXUSA
| | - Alyssa H. Hasty
- Department of Molecular Physiology and BiophysicsVanderbilt University School of MedicineNashvilleTN
- VA Tennessee Valley Healthcare SystemNashvilleTN
| | - Bertha A. Hidalgo
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamAL
| | - Sangwon F. Kim
- Department of Medicine, Division of Endocrinology, Diabetes and MetabolismThe Johns Hopkins University School of MedicineBaltimoreMD
- Department of NeuroscienceThe Johns Hopkins University School of MedicineBaltimoreMD
| | - Julie L. Locher
- Division of Gerontology, Department of Medicine, Geriatrics, and Palliative CareUniversity of Alabama at BirminghamBirminghamAL
| | - James M. Luther
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical Center TennesseeNashvilleTN
| | - Nisa M. Maruthur
- Division of General Internal Medicine, Department of MedicineThe Johns Hopkins University School of MedicineBaltimoreMD
- Department of EpidemiologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchThe Johns Hopkins UniversityBaltimoreMD
| | - Edgar R. Miller
- Division of General Internal Medicine, Department of MedicineThe Johns Hopkins University School of MedicineBaltimoreMD
- Department of EpidemiologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchThe Johns Hopkins UniversityBaltimoreMD
| | - Mary Ann Sevick
- Division of Endocrinology, Department of Medicine, Diabetes and MetabolismNew York University Grossman School of MedicineNew YorkNY
- Department of Population Health, Center for Healthful Behavior ChangeNew York University Langone HealthNew YorkNY
| | - Quinn Wells
- Department of PharmacologyVanderbilt UniversityNashvilleTN
- Department of MedicineVanderbilt University Medical CenterNashvilleTN
| |
Collapse
|
31
|
Dwivedi S, Gottipati A, Ganugula R, Arora M, Friend R, Osburne R, Rodrigues-Hoffman A, Basu R, Pan HL, Kumar MNVR. Oral Nanocurcumin Alone or in Combination with Insulin Alleviates STZ-Induced Diabetic Neuropathy in Rats. Mol Pharm 2022; 19:4612-4624. [PMID: 36106748 PMCID: PMC9972482 DOI: 10.1021/acs.molpharmaceut.2c00465] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus (DM), a multifaceted metabolic disorder if not managed properly leads to secondary complications. Diabetic peripheral neuropathy (DPN) is one such complication caused by nerve damage that cannot be reversed but can be delayed. Recently, diabetes patients are using dietary supplements, although there remains a general skepticism about this practice. Curcumin (CUR), one such supplement can help prevent underlying low-grade inflammation in diabetes, but it is plagued by poor oral bioavailability. To better understand the role of bioavailability in clinical outcomes, we have tested double-headed nanosystems containing curcumin (nCUR) on DPN. Because CUR does not influence glucose levels, we have also tested the effects of nCUR combined with long-acting subcutaneous insulin (INS). nCUR with or without INS alleviates DPN at two times lower dose than unformulated CUR, as indicated by qualitative and quantitative analysis of the hind paw, sciatic nerve, spleen, and L4-6 spinal cord. In addition, nCUR and nCUR+INS preserve hind paw nerve axons as evident by the Bielschowsky silver stain and intraepidermal nerve fibers (IENF) density measured by immunofluorescence. The mechanistic studies further corroborated the results, where nCUR or nCUR+INS showed a significant decrease in TUNEL positive cells, mRNA expression of NLRP3, IL-1β, and macrophage infiltration while preserving nestin and NF200 expression in the sciatic nerve. Together, the data confirms that CUR bioavailability is proportional to clinical outcomes and INS alone may not be one of the solutions for DM. This study highlights the potential of nCUR with or without INS in alleviating DPN and warrants further investigation.
Collapse
Affiliation(s)
- Subhash Dwivedi
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
| | - Anuhya Gottipati
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
| | - Raghu Ganugula
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
| | - Meenakshi Arora
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
| | - Richard Friend
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
| | - Robert Osburne
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
| | - Aline Rodrigues-Hoffman
- Department of Comparative, Diagnostic & Population Medicine, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, Florida32611-7011, United States
| | - Rita Basu
- Division of Endocrinology, Center of Diabetes Technology, University of Virginia School of Medicine, Charlottesville, Virginia22908, United States
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas77030, United States
| | - M N V Ravi Kumar
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama35487-0166, United States
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas77843, United States
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama35401, United States
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama35401, United States
| |
Collapse
|
32
|
Zheng W, Li H, Go Y, Chan XH(F, Huang Q, Wu J. Research Advances on the Damage Mechanism of Skin Glycation and Related Inhibitors. Nutrients 2022; 14:4588. [PMID: 36364850 PMCID: PMC9655929 DOI: 10.3390/nu14214588] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 07/30/2023] Open
Abstract
Our skin is an organ with the largest contact area between the human body and the external environment. Skin aging is affected directly by both endogenous factors and exogenous factors (e.g., UV exposure). Skin saccharification, a non-enzymatic reaction between proteins, e.g., dermal collagen and naturally occurring reducing sugars, is one of the basic root causes of endogenous skin aging. During the reaction, a series of complicated glycation products produced at different reaction stages and pathways are usually collectively referred to as advanced glycation end products (AGEs). AGEs cause cellular dysfunction through the modification of intracellular molecules and accumulate in tissues with aging. AGEs are also associated with a variety of age-related diseases, such as diabetes, cardiovascular disease, renal failure (uremia), and Alzheimer's disease. AGEs accumulate in the skin with age and are amplified through exogenous factors, e.g., ultraviolet radiation, resulting in wrinkles, loss of elasticity, dull yellowing, and other skin problems. This article focuses on the damage mechanism of glucose and its glycation products on the skin by summarizing the biochemical characteristics, compositions, as well as processes of the production and elimination of AGEs. One of the important parts of this article would be to summarize the current AGEs inhibitors to gain insight into the anti-glycation mechanism of the skin and the development of promising natural products with anti-glycation effects.
Collapse
Affiliation(s)
- Wenge Zheng
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, Nanjing 210009, China
| | - Huijuan Li
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, Nanjing 210009, China
| | - Yuyo Go
- Royal Victoria Hospital, BT12 6BA Belfast, Northern Ireland, UK
| | | | - Qing Huang
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, Nanjing 210009, China
| | - Jianxin Wu
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
33
|
Singh H, Agrawal DK. Therapeutic Potential of Targeting the HMGB1/RAGE Axis in Inflammatory Diseases. Molecules 2022; 27:7311. [PMID: 36364135 PMCID: PMC9658169 DOI: 10.3390/molecules27217311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 10/18/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a nuclear protein that can interact with a receptor for advanced glycation end-products (RAGE; a multi-ligand immunoglobulin receptor) and mediates the inflammatory pathways that lead to various pathological conditions, such as cancer, diabetes, neurodegenerative disorders, and cardiovascular diseases. Blocking the HMGB1/RAGE axis could be an effective therapeutic approach to treat these inflammatory conditions, which has been successfully employed by various research groups recently. In this article, we critically review the structural insights and functional mechanism of HMGB1 and RAGE to mediate inflammatory processes. More importantly, current perspectives of recent therapeutic approaches utilized to inhibit the communication between HMGB1 and RAGE using small molecules are also summarized along with their clinical progression to treat various inflammatory disorders. Encouraging results are reported by investigators focusing on HMGB1/RAGE signaling leading to the identification of compounds that could be useful in further clinical studies. We highlight the current gaps in our knowledge and future directions for the therapeutic potential of targeting key molecules in HMGB1/RAGE signaling in the pathophysiology of inflammatory diseases.
Collapse
Affiliation(s)
| | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
34
|
Rojas A, Lindner C, Schneider I, Gonzàlez I, Morales MA. Receptor of advanced glycation end-products axis and gallbladder cancer: A forgotten connection that we should reconsider. World J Gastroenterol 2022; 28:5679-5690. [PMID: 36338887 PMCID: PMC9627425 DOI: 10.3748/wjg.v28.i39.5679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/05/2022] [Accepted: 09/09/2022] [Indexed: 02/06/2023] Open
Abstract
Compelling evidence derived from clinical and experimental research has demonstrated the crucial contribution of chronic inflammation in the development of neoplasms, including gallbladder cancer. In this regard, data derived from clinical and experimental studies have demonstrated that the receptor of advanced glycation end-products (RAGE)/AGEs axis plays an important role in the onset of a crucial and long-lasting inflammatory milieu, thus supporting tumor growth and development. AGEs are formed in biological systems or foods, and food-derived AGEs, also known as dietary AGEs are known to contribute to the systemic pool of AGEs. Once they bind to RAGE, the activation of multiple and crucial signaling pathways are triggered, thus favoring the secretion of several proinflammatory cytokines also involved in the promotion of gallbladder cancer invasion and migration. In the present review, we aimed to highlight the relevance of the association between high dietary AGEs intakes and high risk for gallbladder cancer, and emerging data supporting that dietary intervention to reduce gallbladder cancer risk is a very attractive approach that deserves much more research efforts.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Laboratories, Catholic University of Maule, Talca 34600000, Maule, Chile
| | - Cristian Lindner
- Medicine Faculty, Catholic University of Maule, Talca 34600000, Maule, Chile
| | - Iván Schneider
- Medicine Faculty, Catholic University of Maule, Talca 34600000, Maule, Chile
| | - Ileana Gonzàlez
- Biomedical Research Laboratories, Catholic University of Maule, Talca 34600000, Maule, Chile
| | - Miguel Angel Morales
- Department of Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago 8320000, Santiago, Chile
| |
Collapse
|
35
|
Curran CS, Kopp JB. RAGE pathway activation and function in chronic kidney disease and COVID-19. Front Med (Lausanne) 2022; 9:970423. [PMID: 36017003 PMCID: PMC9395689 DOI: 10.3389/fmed.2022.970423] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/21/2022] [Indexed: 12/23/2022] Open
Abstract
The multi-ligand receptor for advanced glycation end-products (RAGE) and its ligands are contributing factors in autoimmunity, cancers, and infectious disease. RAGE activation is increased in chronic kidney disease (CKD) and coronavirus disease 2019 (COVID-19). CKD may increase the risk of COVID-19 severity and may also develop in the form of long COVID. RAGE is expressed in essentially all kidney cell types. Increased production of RAGE isoforms and RAGE ligands during CKD and COVID-19 promotes RAGE activity. The downstream effects include cellular dysfunction, tissue injury, fibrosis, and inflammation, which in turn contribute to a decline in kidney function, hypertension, thrombotic disorders, and cognitive impairment. In this review, we discuss the forms and mechanisms of RAGE and RAGE ligands in the kidney and COVID-19. Because various small molecules antagonize RAGE activity in animal models, targeting RAGE, its co-receptors, or its ligands may offer novel therapeutic approaches to slowing or halting progressive kidney disease, for which current therapies are often inadequate.
Collapse
Affiliation(s)
- Colleen S. Curran
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey B. Kopp
- Kidney Disease Section, NIDDK (National Institute of Diabetes and Digestive and Kidney Diseases), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
36
|
Dong H, Zhang Y, Huang Y, Deng H. Pathophysiology of RAGE in inflammatory diseases. Front Immunol 2022; 13:931473. [PMID: 35967420 PMCID: PMC9373849 DOI: 10.3389/fimmu.2022.931473] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a non-specific multi-ligand pattern recognition receptor capable of binding to a range of structurally diverse ligands, expressed on a variety of cell types, and performing different functions. The ligand-RAGE axis can trigger a range of signaling events that are associated with diabetes and its complications, neurological disorders, cancer, inflammation and other diseases. Since RAGE is involved in the pathophysiological processes of many diseases, targeting RAGE may be an effective strategy to block RAGE signaling.
Collapse
|
37
|
Arivazhagan L, López-Díez R, Shekhtman A, Ramasamy R, Schmidt AM. Glycation and a Spark of ALEs (Advanced Lipoxidation End Products) - Igniting RAGE/Diaphanous-1 and Cardiometabolic Disease. Front Cardiovasc Med 2022; 9:937071. [PMID: 35811725 PMCID: PMC9263181 DOI: 10.3389/fcvm.2022.937071] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/30/2022] [Indexed: 12/25/2022] Open
Abstract
Obesity and non-alcoholic fatty liver disease (NAFLD) are on the rise world-wide; despite fervent advocacy for healthier diets and enhanced physical activity, these disorders persist unabated and, long-term, are major causes of morbidity and mortality. Numerous fundamental biochemical and molecular pathways participate in these events at incipient, mid- and advanced stages during atherogenesis and impaired regression of established atherosclerosis. It is proposed that upon the consumption of high fat/high sugar diets, the production of receptor for advanced glycation end products (RAGE) ligands, advanced glycation end products (AGEs) and advanced lipoxidation end products (ALEs), contribute to the development of foam cells, endothelial injury, vascular inflammation, and, ultimately, atherosclerosis and its consequences. RAGE/Diaphanous-1 (DIAPH1) increases macrophage foam cell formation; decreases cholesterol efflux and causes foam cells to produce and release damage associated molecular patterns (DAMPs) molecules, which are also ligands of RAGE. DAMPs stimulate upregulation of Interferon Regulatory Factor 7 (IRF7) in macrophages, which exacerbates vascular inflammation and further perturbs cholesterol metabolism. Obesity and NAFLD, characterized by the upregulation of AGEs, ALEs and DAMPs in the target tissues, contribute to insulin resistance, hyperglycemia and type two diabetes. Once in motion, a vicious cycle of RAGE ligand production and exacerbation of RAGE/DIAPH1 signaling ensues, which, if left unchecked, augments cardiometabolic disease and its consequences. This Review focuses on RAGE/DIAPH1 and its role in perturbation of metabolism and processes that converge to augur cardiovascular disease.
Collapse
Affiliation(s)
- Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY, United States
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States,*Correspondence: Ann Marie Schmidt
| |
Collapse
|
38
|
Ramasamy R, Shekhtman A, Schmidt AM. The RAGE/DIAPH1 Signaling Axis & Implications for the Pathogenesis of Diabetic Complications. Int J Mol Sci 2022; 23:ijms23094579. [PMID: 35562970 PMCID: PMC9102165 DOI: 10.3390/ijms23094579] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/13/2022] [Accepted: 04/17/2022] [Indexed: 02/08/2023] Open
Abstract
Increasing evidence links the RAGE (receptor for advanced glycation end products)/DIAPH1 (Diaphanous 1) signaling axis to the pathogenesis of diabetic complications. RAGE is a multi-ligand receptor and through these ligand-receptor interactions, extensive maladaptive effects are exerted on cell types and tissues targeted for dysfunction in hyperglycemia observed in both type 1 and type 2 diabetes. Recent evidence indicates that RAGE ligands, acting as damage-associated molecular patterns molecules, or DAMPs, through RAGE may impact interferon signaling pathways, specifically through upregulation of IRF7 (interferon regulatory factor 7), thereby heralding and evoking pro-inflammatory effects on vulnerable tissues. Although successful targeting of RAGE in the clinical milieu has, to date, not been met with success, recent approaches to target RAGE intracellular signaling may hold promise to fill this critical gap. This review focuses on recent examples of highlights and updates to the pathobiology of RAGE and DIAPH1 in diabetic complications.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY 12222, USA;
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA;
- Correspondence:
| |
Collapse
|
39
|
Huang Y, Xin W, Xiong J, Yao M, Zhang B, Zhao J. The Intestinal Microbiota and Metabolites in the Gut-Kidney-Heart Axis of Chronic Kidney Disease. Front Pharmacol 2022; 13:837500. [PMID: 35370631 PMCID: PMC8971625 DOI: 10.3389/fphar.2022.837500] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Emerging evidences demonstrate the involvement of gut microbiota in the progression of chronic kidney disease (CKD) and CKD-associated complications including cardiovascular disease (CVD) and intestinal dysfunction. In this review, we discuss the interactions between the gut, kidney and heart in CKD state, and elucidate the significant role of intestinal microbiota in the gut-kidney-heart axis hypothesis for the pathophysiological mechanisms of these diseases, during which process mitochondria may serve as a potential therapeutic target. Dysregulation of this axis will lead to a vicious circle, contributing to CKD progression. Recent studies suggest novel therapies targeting gut microbiota in the gut-kidney-heart axis, including dietary intervention, probiotics, prebiotics, genetically engineered bacteria, fecal microbiota transplantation, bacterial metabolites modulation, antibiotics, conventional drugs and traditional Chinese medicine. Further, the identification of specific microbial communities and their corresponding pathophysiological metabolites and the illumination of the gut-kidney-heart axis may contribute to innovative basic research, clinical trials and therapeutic strategies against CKD progression and uremic complications in CKD patients.
Collapse
|
40
|
|