1
|
Jones TLZ, Kusmartseva I, Litovsky S, Thakar R, Posgai AL, Eckel RH, Atkinson MA. The Cardiovascular Repository for Type 1 Diabetes (CaRe-T1D): An NIDDK Initiative to Advance Understanding of Mechanisms Underlying Cardiovascular Disease in Type 1 Versus Type 2 Diabetes. Diabetes 2025; 74:1078-1088. [PMID: 40272262 DOI: 10.2337/db25-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality in individuals with diabetes. Individuals with type 1 diabetes have a two- to fourfold higher risk of CVD in comparison with the general population, driven by an earlier onset and increased lifetime incidence of CVD events and mortality. Similarly, type 2 diabetes confers two- to threefold increased CVD risk, usually alongside metabolic syndrome, obesity, and hypertension. Despite advancements in methods for achieving glycemic control, the CVD burden remains disproportionately high in diabetes. The mechanisms driving elevated risk are complex and variably multifactorial, involving hyperglycemia, insulin resistance, dyslipidemia, inflammation, and a hypercoagulable state. Unfortunately, critical gaps in understanding persist on how these factors interact to promote CVD in type 1 versus type 2 diabetes, particularly across disease stages and age. Addressing these knowledge gaps is essential to developing targeted therapies that can effectively mitigate CVD risk. To meet this need, the National Institute of Diabetes and Digestive and Kidney Diseases, in partnership with the National Heart, Lung, and Blood Institute, recently formed the Cardiovascular Repository for Type 1 Diabetes (CaRe-T1D) program. Its mission is to elucidate the molecular and cellular pathways linking diabetes with CVD through the provision of high-quality human tissues for investigator-led analyses using cutting-edge technologies and collaborative data sharing to advance precision medicine and reduce the global burden of diabetes-associated cardiovascular complications. ARTICLE HIGHLIGHTS CaRe-T1D established a biorepository and scientific consortium to advance research on cardiovascular complications in diabetes. The goal is to determine how cardiovascular disease differs in type 1 versus type 2 diabetes. Heart, kidney, carotid and peripheral arteries, and blood from organ donors with type 1 diabetes, with type 2 diabetes, or without diabetes will be distributed to approved investigators to address the pathogenesis of diabetic cardiovascular disease. CaRe-T1D is a resource of human cardiovascular tissue and a database with the results from tissue analysis.
Collapse
Affiliation(s)
- Teresa L Z Jones
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Irina Kusmartseva
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
| | - Silvio Litovsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Rahul Thakar
- National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
| | - Robert H Eckel
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
2
|
Huang K, Mi B, Xiong Y, Fu Z, Zhou W, Liu W, Liu G, Dai G. Angiogenesis during diabetic wound repair: from mechanism to therapy opportunity. BURNS & TRAUMA 2025; 13:tkae052. [PMID: 39927093 PMCID: PMC11802347 DOI: 10.1093/burnst/tkae052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/23/2024] [Accepted: 08/09/2024] [Indexed: 02/11/2025]
Abstract
Diabetes mellitus, a pervasive chronic metabolic disorder, is often associated with complications such as impaired wound healing. Various factors, most notably vascular deficiency, govern the wound repair process in diabetic patients, significantly impeding diabetic wound healing; therefore, angiogenesis and its role in diabetic wound repair have emerged as important areas of research. This review aims to delve into the mechanisms of angiogenesis, the effects of diabetes on angiogenesis, and the association between angiogenesis and diabetic wound repair. This will ultimately offer valuable guidance regarding the ideal timing of diabetic wound treatment in a clinical setting.
Collapse
Affiliation(s)
- Kang Huang
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Bobin Mi
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Yuan Xiong
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Zicai Fu
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Wenyun Zhou
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Wanjun Liu
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Guohui Liu
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Guandong Dai
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| |
Collapse
|
3
|
Erim B, Binici Hİ. Advanced glycation end products: understanding their health risks and effective prevention strategies. NUTRIRE 2024; 49:54. [DOI: 10.1186/s41110-024-00298-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 01/03/2025]
|
4
|
Larry M, Rabizadeh S, Mohammadi F, Yadegar A, Jalalpour A, Mirmiranpour H, Farahmand G, Esteghamati A, Nakhjavani M. Relationship between advanced glycation end-products and advanced oxidation protein products in patients with type 2 diabetes with and without albuminuria: A cross-sectional survey. Health Sci Rep 2024; 7:e70057. [PMID: 39355098 PMCID: PMC11439888 DOI: 10.1002/hsr2.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 10/03/2024] Open
Abstract
Background and Aims Literature suggests that oxidative stress plays a crucial role in the progression of diabetes. Since poor glycemic control enhances the formation of advanced glycation end-products (AGEs) and advanced oxidation protein products (AOPP) in individuals with diabetes, exploring the association between glycation and oxidative states in diabetes could also shed light on potential consequences. This study evaluated the effects of albuminuria on AGEs and AOPP levels and measured their relationship in participants with type 2 diabetes (T2D) with or without albuminuria. Methods A cross-sectional, matched case-control study was designed, including 38 T2D subjects with albuminuria and 38 matched T2D subjects with normoalbuminuria. Patients were matched by their body mass index (BMI), age, and duration of diabetes. The unadjusted and adjusted correlation between AGEs and AOPP in the studied groups were analyzed by multiple logistic regression. Using ggplot2, the ties between these two biochemical factors in cases and controls were plotted. Results This study elucidated a significant association between AGEs and AOPP in participants with normoalbuminuria (r = 0.331, p-value < 0.05), which continued to be significant after controlling for BMI, age, systolic blood pressure (SBP), and diastolic blood pressure (DBP) (r = 0.355, p-value < 0.05). However, there was no significant association between AGEs and AOPP in those with albuminuria in the unadjusted model (r = 0.034, p-value = 0.841) or after controlling for BMI, age, SBP, and DBP (r = 0.076, p-value = 0.685). Conclusion Oxidation and glycation molecular biomarkers were correlated in patients without albuminuria; however, this association was not observed in those with albuminuria.
Collapse
Affiliation(s)
- Mehrdad Larry
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Soghra Rabizadeh
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Fatemeh Mohammadi
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Amirhossein Yadegar
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Azadeh Jalalpour
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Hossein Mirmiranpour
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Ghasem Farahmand
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Alireza Esteghamati
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| | - Manouchehr Nakhjavani
- Endocrinology and Metabolism Research Center (EMRC), Vali‐Asr HospitalTehran University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Geng XF, Shang WY, Qi ZW, Zhang C, Li WX, Yan ZP, Fan XB, Zhang JP. The mechanism and promising therapeutic strategy of diabetic cardiomyopathy dysfunctions: Focus on pyroptosis. J Diabetes Complications 2024; 38:108848. [PMID: 39178624 DOI: 10.1016/j.jdiacomp.2024.108848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
Diabetes is a major risk factor for cardiovascular diseases, and myocardial damage caused by hyperglycemia is the main cause of heart failure. However, there is still a lack of systematic understanding of myocardial damage caused by diabetes. At present, we believe that the cellular inflammatory damage caused by hyperglycemia is one of the causes of diabetic cardiomyopathy. Pyroptosis, as a proinflammatory form of cell death, is closely related to the occurrence and development of diabetic cardiomyopathy. Therefore, this paper focuses on the important role of inflammation in the occurrence and development of diabetic cardiomyopathy. From the perspective of pyroptosis, we summarize the pyroptosis of different types of cells in diabetic cardiomyopathy and its related signaling pathways. It also summarizes the treatment of diabetic cardiomyopathy, hoping to provide methods for the prevention and treatment of diabetic cardiomyopathy by inhibiting pyroptosis.
Collapse
Affiliation(s)
- Xiao-Fei Geng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Wen-Yu Shang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhong-Wen Qi
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Chi Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Wen-Xiu Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhi-Peng Yan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xin-Biao Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Jun-Ping Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
6
|
Tecce N, de Alteriis G, de Alteriis G, Verde L, Tecce MF, Colao A, Muscogiuri G. Harnessing the Synergy of SGLT2 Inhibitors and Continuous Ketone Monitoring (CKM) in Managing Heart Failure among Patients with Type 1 Diabetes. Healthcare (Basel) 2024; 12:753. [PMID: 38610175 PMCID: PMC11011472 DOI: 10.3390/healthcare12070753] [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: 02/20/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Heart failure (HF) management in type 1 diabetes (T1D) is particularly challenging due to its increased prevalence and the associated risks of hospitalization and mortality, driven by diabetic cardiomyopathy. Sodium-glucose cotransporter-2 inhibitors (SGLT2-is) offer a promising avenue for treating HF, specifically the preserved ejection fraction variant most common in T1D, but their utility is hampered by the risk of euglycemic diabetic ketoacidosis (DKA). This review investigates the potential of SGLT2-is in T1D HF management alongside emergent Continuous Ketone Monitoring (CKM) technology as a means to mitigate DKA risk through a comprehensive analysis of clinical trials, observational studies, and reviews. The evidence suggests that SGLT2-is significantly reduce HF hospitalization and enhance cardiovascular outcomes. However, their application in T1D patients remains limited due to DKA concerns. CKM technology emerges as a crucial tool in this context, offering real-time monitoring of ketone levels, which enables the safe incorporation of SGLT2-is into treatment regimes by allowing for early detection and intervention in the development of ketosis. The synergy between SGLT2-is and CKM has the potential to revolutionize HF treatment in T1D, promising improved patient safety, quality of life, and reduced HF-related morbidity and mortality. Future research should aim to employ clinical trials directly assessing this integrated approach, potentially guiding new management protocols for HF in T1D.
Collapse
Affiliation(s)
- Nicola Tecce
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Napoli, Italy; (G.d.A.); (A.C.)
| | - Giorgio de Alteriis
- Department of Industrial Engineering, University of Naples Federico II, Piazzale Tecchio 80, 80125 Naples, Italy;
| | - Giulia de Alteriis
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Napoli, Italy; (G.d.A.); (A.C.)
| | - Ludovica Verde
- Centro Italiano per la Cura e il Benessere del Paziente con Obesità (C.I.B.O), Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Napoli, Italy;
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy;
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Napoli, Italy; (G.d.A.); (A.C.)
- Cattedra Unesco “Educazione alla Salute e Allo Sviluppo Sostenibile”, University Federico II, 80131 Napoli, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Napoli, Italy; (G.d.A.); (A.C.)
- Cattedra Unesco “Educazione alla Salute e Allo Sviluppo Sostenibile”, University Federico II, 80131 Napoli, Italy
| |
Collapse
|
7
|
Zambre S, Bangar N, Mistry A, Katarmal P, Khan MS, Ahmed I, Tupe R, Roy B. Aldosterone, Methylglyoxal, and Glycated Albumin Interaction with Macrophage Cells Affects Their Viability, Activation, and Differentiation. ACS OMEGA 2024; 9:11848-11859. [PMID: 38497023 PMCID: PMC10938338 DOI: 10.1021/acsomega.3c09420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND The inflammatory response in diabetes is strongly correlated with increasing amounts of advanced glycation end products (AGEs), methylglyoxal (MGO), aldosterone (Aldo), and activation of macrophages. Aldo is known to be associated with increased pro-inflammatory responses in general, but its significance in inflammatory responses under glycated circumstances has yet to be understood. In the current work, the aim of our study was to study the macrophage immune response in the presence of AGEs, MGO, and Aldo to comprehend their combined impact on diabetes-associated complications. METHODS AND RESULTS The viability of macrophages upon treatment with glycated HSA (Gly-HSA) promoted cell growth as the concentration increased from 100 to 500 μg/mL, whereas MGO at a high concentration (≥300 μM) significantly hampered cell growth. At lower concentrations (0.5-5 nM), Aldo strongly promoted cell growth, whereas at higher concentrations (50 nM), it was seen to inhibit growth when used for cell treatment for 24 h. Aldo had no effect on MGO-induced cell growth inhibition after 24 h of treatment. However, compared to MGO or Aldo treatment alone, an additional decrease in viability could be seen after 48 h of treatment with a combination of MGO and Aldo. Treatment with Aldo and MGO induced expression of TNF-α independently and when combined. However, when combined, Aldo and MGO significantly suppressed the expression of TGF-β. Aldo, Gly-HSA, and MGO strongly induced the transcription of NF-κB and RAGE mRNA and, as expected, also promoted the formation of reactive oxygen species. Also, by inducing iNOS and MHC-II and suppressing CD206 transcript expression, Gly-HSA strongly favored the differentiation of macrophages into M1 type (pro-inflammatory). On the other hand, the combination of Aldo and MGO strongly induced the expression of MHC-II, CD206, and ARG1 (M2 macrophage marker). These findings suggest that Gly-HSA, MGO, and Aldo differently influence macrophage survival, activation, and differentiation. CONCLUSIONS Overall, this study gives an insight into the effects of glycated protein and MGO in the presence of Aldo on macrophage survival, activation, differentiation, and inflammatory response.
Collapse
Affiliation(s)
- Saee Zambre
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Nilima Bangar
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Armaan Mistry
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Poonam Katarmal
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Mohd Shahnawaz Khan
- Department
of Biochemistry, College of Science, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Irshad Ahmed
- Department
of Biochemistry and Structural Biology, School of Medicine, UT Health Science Center, San Antonio, Texas 78229, United States
| | - Rashmi Tupe
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Bishnudeo Roy
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| |
Collapse
|
8
|
Das S, Devi Rajeswari V, Venkatraman G, Elumalai R, Dhanasekaran S, Ramanathan G. Current updates on metabolites and its interlinked pathways as biomarkers for diabetic kidney disease: A systematic review. Transl Res 2024; 265:71-87. [PMID: 37952771 DOI: 10.1016/j.trsl.2023.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Diabetic kidney disease (DKD) is a major microvascular complication of diabetes mellitus (DM) that poses a serious risk as it can lead to end-stage renal disease (ESRD). DKD is linked to changes in the diversity, composition, and functionality of the microbiota present in the gastrointestinal tract. The interplay between the gut microbiota and the host organism is primarily facilitated by metabolites generated by microbial metabolic processes from both dietary substrates and endogenous host compounds. The production of numerous metabolites by the gut microbiota is a crucial factor in the pathogenesis of DKD. However, a comprehensive understanding of the precise mechanisms by which gut microbiota and its metabolites contribute to the onset and progression of DKD remains incomplete. This review will provide a summary of the current scenario of metabolites in DKD and the impact of these metabolites on DKD progression. We will discuss in detail the primary and gut-derived metabolites in DKD, and the mechanisms of the metabolites involved in DKD progression. Further, we will address the importance of metabolomics in helping identify potential DKD markers. Furthermore, the possible therapeutic interventions and research gaps will be highlighted.
Collapse
Affiliation(s)
- Soumik Das
- School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - V Devi Rajeswari
- School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Ganesh Venkatraman
- School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Ramprasad Elumalai
- Department of Nephrology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu 600116, India
| | - Sivaraman Dhanasekaran
- School of Energy Technology, Pandit Deendayal Energy University, Knowledge Corridor, Raisan Village, PDPU Road, Gandhinagar, Gujarat 382426, India
| | - Gnanasambandan Ramanathan
- School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
9
|
Dobrucki IT, Miskalis A, Nelappana M, Applegate C, Wozniak M, Czerwinski A, Kalinowski L, Dobrucki LW. Receptor for advanced glycation end-products: Biological significance and imaging applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1935. [PMID: 37926944 DOI: 10.1002/wnan.1935] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
The receptor for advanced glycation end-products (RAGE or AGER) is a transmembrane, immunoglobulin-like receptor that, due to its multiple isoform structures, binds to a diverse range of endo- and exogenous ligands. RAGE activation caused by the ligand binding initiates a cascade of complex pathways associated with producing free radicals, such as reactive nitric oxide and oxygen species, cell proliferation, and immunoinflammatory processes. The involvement of RAGE in the pathogenesis of disorders such as diabetes, inflammation, tumor progression, and endothelial dysfunction is dictated by the accumulation of advanced glycation end-products (AGEs) at pathologic states leading to sustained RAGE upregulation. The involvement of RAGE and its ligands in numerous pathologies and diseases makes RAGE an interesting target for therapy focused on the modulation of both RAGE expression or activation and the production or exogenous administration of AGEs. Despite the known role that the RAGE/AGE axis plays in multiple disease states, there remains an urgent need to develop noninvasive, molecular imaging approaches that can accurately quantify RAGE levels in vivo that will aid in the validation of RAGE and its ligands as biomarkers and therapeutic targets. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Iwona T Dobrucki
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Academy of Medical and Social Applied Sciences, Elblag, Poland
| | - Angelo Miskalis
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Michael Nelappana
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Catherine Applegate
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Cancer Center at Illinois, Urbana, Illinois, USA
| | - Marcin Wozniak
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Division of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
| | - Andrzej Czerwinski
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Leszek Kalinowski
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Division of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | - Lawrence W Dobrucki
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, Urbana, Illinois, USA
- Division of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
10
|
Xu CM, Karbasiafshar C, Brinck‐Teixeira R, Broadwin M, Sellke FW, Abid MR. Diabetic state of human coronary artery endothelial cells results in altered effects of bone mesenchymal stem cell-derived extracellular vesicles. Physiol Rep 2023; 11:e15866. [PMID: 38114067 PMCID: PMC10730301 DOI: 10.14814/phy2.15866] [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: 07/10/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 12/21/2023] Open
Abstract
Human bone mesenchymal stem cell-derived extracellular vesicles (HBMSC-EV) have been used successfully in animal models of myocardial ischemia, yet have dampened effects in metabolic syndrome through unknown mechanisms. This study demonstrates the basal differences between non-diabetic human coronary artery endothelial cells (HCAEC) and diabetic HCAEC (DM-HCAEC), and how these cells respond to the treatment of HBMSC-EV. HCAEC and DM-HCAEC were treated with HBMSC-EV for 6 h. Proteomics, western blot analysis, and tube formation assays were performed. Key metabolic, growth, and stress/starvation cellular responses were significantly altered in DM-HCAEC in comparison to that of HCAEC at baseline. Proteomics demonstrated increased phosphorus metabolic process and immune pathways and decreased RNA processing and biosynthetic pathways in DM-HCAEC. Similar to previous in vivo findings, HCAEC responded to the HBMSC-EV with regenerative and anti-inflammatory effects through the upregulation of multiple RNA pathways and downregulation of immune cell activation pathways. In contrast, DM-HCAEC had a significantly diminished response to HBMSC-EV, likely due to the baseline abnormalities in DM-HCAEC. To achieve the full benefits of HBMSC-EV and for a successful transition of this potential therapeutic agent to clinical studies, the abnormalities found in DM-HCAEC will need to be further studied.
Collapse
Affiliation(s)
- Cynthia M. Xu
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | | | - Rayane Brinck‐Teixeira
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | - Mark Broadwin
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | - Frank W. Sellke
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| | - M. Ruhul Abid
- Cardiovascular Research Center, Rhode Island HospitalProvidenceRhode IslandUSA
- Division of Cardiothoracic SurgeryAlpert Medical School of Brown University and Rhode Island HospitalProvidenceRhode IslandUSA
| |
Collapse
|
11
|
Chen M, Hu J, Chen C, Hao G, Hu S, Xu J, Hu C. Radiomics analysis of pericoronary adipose tissue based on plain CT for preliminary screening of coronary artery disease in patients with type 2 diabetes mellitus. Acta Radiol 2023; 64:2704-2713. [PMID: 37603886 DOI: 10.1177/02841851231189998] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is associated with a markedly increased prevalence of coronary artery disease (CAD). Radiomics features of pericoronary adipose tissue (PCAT) were correlated with inflammation, which may have potential value in the prediction of CAD. PURPOSE To determine whether radiomics analysis of PCAT captured by plain computed tomography (CT) could predict obstructive CAD in patients with T2DM. MATERIAL AND METHODS The study included 155 patients with T2DM with suspected CAD between January 2020 and December 2021. Volumes of right coronary artery of 10-50 mm were delineated in the plain CT to extract radiomics features and PCAT CT attenuation (PCATa). Least absolute shrinkage and selection operator was used to select the useful radiomics features to calculate the radiomics score (Rad-score). Univariate and multivariable logistic regression were applied to select independent predictors. The predictive performance was evaluated by the area under the receiver operating characteristics curve (AUC). RESULTS Rad-score (per 0.1 increments: odds ratio [OR] = 1.297; P < 0.001), coronary artery calcium score (CACS) (OR = 1.003; P = 0.037), and sex (OR = 3.245; P = 0.026) were identified as independent predictors for obstructive CAD. Rad-score (AUC = 0.835) outperformed CACS (AUC = 0.780), sex (AUC = 0.665), and PCATa (AUC = 0.550) in predicting obstructive CAD (P = 0.017 and 0.003 for Rad-score vs. sex and PCATa, respectively); however, the improvement between Rad-score and CACS had no statistical significance (P = 0.490). CONCLUSION Plain CT-derived Rad-score may be used as a preliminary screening tool for obstructive CAD in patients with T2DM.
Collapse
Affiliation(s)
- Meng Chen
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Medical Imaging, Soochow University, Suzhou, PR China
| | - Jingcheng Hu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Can Chen
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Medical Imaging, Soochow University, Suzhou, PR China
| | - Guangyu Hao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Medical Imaging, Soochow University, Suzhou, PR China
| | - Su Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Medical Imaging, Soochow University, Suzhou, PR China
| | - Jialiang Xu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Medical Imaging, Soochow University, Suzhou, PR China
| |
Collapse
|
12
|
Bansal S, Burman A, Tripathi AK. Advanced glycation end products: Key mediator and therapeutic target of cardiovascular complications in diabetes. World J Diabetes 2023; 14:1146-1162. [PMID: 37664478 PMCID: PMC10473940 DOI: 10.4239/wjd.v14.i8.1146] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/21/2023] [Accepted: 05/22/2023] [Indexed: 08/11/2023] Open
Abstract
The incidence of type 2 diabetes mellitus is growing in epidemic proportions and has become one of the most critical public health concerns. Cardiovascular complications associated with diabetes are the leading cause of morbidity and mortality. The cardiovascular diseases that accompany diabetes include angina, myocardial infarction, stroke, peripheral artery disease, and congestive heart failure. Among the various risk factors generated secondary to hyperglycemic situations, advanced glycation end products (AGEs) are one of the important targets for future diagnosis and prevention of diabetes. In the last decade, AGEs have drawn a lot of attention due to their involvement in diabetic patho-physiology. AGEs can be derived exogenously and endogenously through various pathways. These are a non-homogeneous, chemically diverse group of compounds formed non-enzymatically by condensation between carbonyl groups of reducing sugars and free amino groups of protein, lipids, and nucleic acid. AGEs mediate their pathological effects at the cellular and extracellular levels by multiple pathways. At the cellular level, they activate signaling cascades via the receptor for AGEs and initiate a complex series of intracellular signaling resulting in reactive oxygen species generation, inflammation, cellular proliferation, and fibrosis that may possibly exacerbate the damaging effects on cardiac functions in diabetics. AGEs also cause covalent modifications and cross-linking of serum and extracellular matrix proteins; altering their structure, stability, and functions. Early diagnosis of diabetes may prevent its progression to complications and decrease its associated comorbidities. In the present review, we recapitulate the role of AGEs as a crucial mediator of hyperglycemia-mediated detrimental effects in diabetes-associated complications. Furthermore, this review presents an overview of future perspectives for new therapeutic interventions to ameliorate cardiovascular complications in diabetes.
Collapse
Affiliation(s)
- Savita Bansal
- Department of Biochemistry, Institute of Home Sciences, University of Delhi, New Delhi 110016, India
| | - Archana Burman
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi 110016, India
| | - Asok Kumar Tripathi
- Department of Biochemistry, University College of Medical Sciences, University of Delhi, New Delhi 110095, India
| |
Collapse
|
13
|
Liu H, Wang X, Gao H, Yang C, Xie C. Physiological and pathological characteristics of vascular endothelial injury in diabetes and the regulatory mechanism of autophagy. Front Endocrinol (Lausanne) 2023; 14:1191426. [PMID: 37441493 PMCID: PMC10333703 DOI: 10.3389/fendo.2023.1191426] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Vascular endothelial injury in diabetes mellitus (DM) is the major cause of vascular disease, which is closely related to the occurrence and development of a series of vascular complications and has a serious negative impact on a patient's health and quality of life. The primary function of normal vascular endothelium is to function as a barrier function. However, in the presence of DM, glucose and lipid metabolism disorders, insulin resistance, inflammatory reactions, oxidative stress, and other factors cause vascular endothelial injury, leading to vascular endothelial lesions from morphology to function. Recently, numerous studies have found that autophagy plays a vital role in regulating the progression of vascular endothelial injury. Therefore, this article compares the morphology and function of normal and diabetic vascular endothelium and focuses on the current regulatory mechanisms and the important role of autophagy in diabetic vascular endothelial injury caused by different signal pathways. We aim to provide some references for future research on the mechanism of vascular endothelial injury in DM, investigate autophagy's protective or injurious effect, and study potential drugs using autophagy as a target.
Collapse
Affiliation(s)
- Hanyu Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xueru Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, China
| | - Chan Yang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
14
|
Sonmez MI, Shahzadi A, Kose C, Sonmez H, Ozyazgan S, Akkan AG. Effect of sulfasalazine on endothelium-dependent vascular response by the activation of Nrf2 signalling pathway. Front Pharmacol 2022; 13:979300. [PMID: 36353481 PMCID: PMC9639785 DOI: 10.3389/fphar.2022.979300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Diabetes mellitus leads to endothelial dysfunction and accumulation of oxygen radicals. Sulfasalazine-induced Nrf2 activation reduces oxidative stress in vessels. Thus, in the present study, we investigated the effects of sulfasalazine on endothelial dysfunction induced by high glucose. We also ascribed the underlying mechanism involved in glucose-induced endothelial dysfunction. Methods: For this experiment we used 80 Wistar Albino rats thoracic aorta to calculate the dose response curve of noradrenaline and acetylcholine. Vessels were incubated in normal and high glucose for 2 h. To investigate glucose and sulfasalazine effects the vessels of the high glucose group were pre-treated with sulfasalazine (300 mM), JNK inhibitor (SP600125), and ERK inhibitor (U0126) for 30 min. The dose response curve was calculated through organ bath. The eNOS, TAS, TOS, and HO-1 levels were estimated by commercially available ELISA kits. Results: In the high glucose group, the Emax for contraction was significantly higher (p < 0.001), and Emax for relaxation was lower than that of control. These functional changes were parallel with the low levels of eNOS (p < 0.05). High glucose vessel treated with sulfasalazine showed low Emax value for contraction (p < 0.001) however, the Emax for relaxation was significantly high (p < 0.001) when compared to high glucose group. In the JNK group, Emax for contraction and relaxation was inhibited (p < 0.001) compared to sulfasalazine treated vessels. HO—1 enzyme levels were significantly low (p < 0.01) with sulfasalazine but higher with ERK inhibitor (p < 0.05). Conclusion: High glucose induced endothelial dysfunction and sulfasalazine reduced damage in high glucose vessels by activating eNOS, antioxidant effect through HO-1 enzymes and particularly inducing Nrf2 via the ERK and JNK pathways.
Collapse
Affiliation(s)
- Muhammed Ikbal Sonmez
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- *Correspondence: Muhammed Ikbal Sonmez,
| | - Andleeb Shahzadi
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Cagla Kose
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
- Department of Medical Pharmacology, Medical Faculty, Halic University, Istanbul, Turkey
| | - Haktan Sonmez
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sibel Ozyazgan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ahmet Gokhan Akkan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
- Department of Medical Pharmacology, Medical Faculty, Bezmialem Vakif University Hospital, Istanbul, Turkey
| |
Collapse
|
15
|
Abudureyimu M, Luo X, Wang X, Sowers JR, Wang W, Ge J, Ren J, Zhang Y. Heart failure with preserved ejection fraction (HFpEF) in type 2 diabetes mellitus: from pathophysiology to therapeutics. J Mol Cell Biol 2022; 14:mjac028. [PMID: 35511596 PMCID: PMC9465638 DOI: 10.1093/jmcb/mjac028] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Accepted: 04/29/2022] [Indexed: 11/30/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM or T2D) is a devastating metabolic abnormality featured by insulin resistance, hyperglycemia, and hyperlipidemia. T2D provokes unique metabolic changes and compromises cardiovascular geometry and function. Meanwhile, T2D increases the overall risk for heart failure (HF) and acts independent of classical risk factors including coronary artery disease, hypertension, and valvular heart diseases. The incidence of HF is extremely high in patients with T2D and is manifested as HF with preserved, reduced, and midrange ejection fraction (HFpEF, HFrEF, and HFmrEF, respectively), all of which significantly worsen the prognosis for T2D. HFpEF is seen in approximately half of the HF cases and is defined as a heterogenous syndrome with discrete phenotypes, particularly in close association with metabolic syndrome. Nonetheless, management of HFpEF in T2D remains unclear, largely due to the poorly defined pathophysiology behind HFpEF. Here, in this review, we will summarize findings from multiple preclinical and clinical studies as well as recent clinical trials, mainly focusing on the pathophysiology, potential mechanisms, and therapies of HFpEF in T2D.
Collapse
Affiliation(s)
- Miyesaier Abudureyimu
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200031, China
| | - Xuanming Luo
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200031, China
| | - Xiang Wang
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200031, China
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri Columbia, Columbia, MO 65212, USA
| | - Wenshuo Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Aftermath of AGE-RAGE Cascade in the pathophysiology of cardiovascular ailments. Life Sci 2022; 307:120860. [PMID: 35940220 DOI: 10.1016/j.lfs.2022.120860] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022]
|
17
|
Choi LS, Ahmed K, Kim YS, Yim JE. Skin accumulation of advanced glycation end products and cardiovascular risk in Korean patients with type 2 diabetes mellitus. Heliyon 2022; 8:e09571. [PMID: 35711980 PMCID: PMC9192809 DOI: 10.1016/j.heliyon.2022.e09571] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/01/2022] [Accepted: 05/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background The formation of advanced glycation end products (AGEs) takes place during normal aging; however, their production is faster in people having diabetes. The accumulated AGEs reportedly play a role in the occurrence of various age-related disorders. Furthermore, the skin autofluorescence (SAF) technique can be used to detect accumulated AGEs levels. There are few reports on the association between skin accumulation of AGEs and risk of complications in type 2 diabetes mellitus. Methods In this study, we aimed to describe the association between the skin accumulation of AGEs and cardiovascular risk factors in Korean patients with type 2 diabetes. A total of 310 Korean patients with diabetes were enrolled, and the levels of AGEs were measured using SAP. Levels of fasting blood glucose (FBS), triglycerides, total cholesterol, low- and high-density lipoprotein cholesterol, proteinuria, arterial pulse wave velocity (PWV), and blood vessel age were measured using an automatic waveform analyzer. General linear models were used to identify the independent effect of AGEs after adjusting for covariates (age, weight, and duration of diabetes). Results The skin levels of AGEs were strongly correlated with the diabetes duration. Significant independent associations were observed for AGEs with FBS (P < 0.01), proteinuria (P < 0.001), and PWV (P < 0.001). The advanced glycated product was independently associated to the arterial pulse wave conduction velocity that is used as a representative method for measuring arteriosclerosis by analysis early cardiovascular risk factors. Conclusion Our results show that an increase in SAF levels in Korean patients with type 2 diabetes is associated with PWV and vein age, and thereby with arterial stiffness. Therefore, our results suggest that AGEs are associated with cardiovascular risk factors. The level of AGEs can thus be used as an indicator of cardiovascular diseases in the clinical diagnosis of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Lee-Seoul Choi
- Department of Food and Nutrition, Changwon National University, Changwon, South Korea
| | - Kainat Ahmed
- Interdisciplinary Program in Senior Human Ecology, Changwon National University, Changwon, South Korea
| | - Young-Seol Kim
- Department of Endocrinology, Kyung Hee Medical Center, Seoul, South Korea
| | - Jung-Eun Yim
- Department of Food and Nutrition, Changwon National University, Changwon, South Korea
- Interdisciplinary Program in Senior Human Ecology, Changwon National University, Changwon, South Korea
| |
Collapse
|
18
|
Singh S, Siva BV, Ravichandiran V. Advanced Glycation End Products: key player of the pathogenesis of atherosclerosis. Glycoconj J 2022; 39:547-563. [PMID: 35579827 DOI: 10.1007/s10719-022-10063-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 01/08/2023]
Abstract
Atherosclerosis is the most common type of cardiovascular disease, and it causes intima thickening, plaque development, and ultimate blockage of the artery lumen. Advanced glycation end products (AGEs) are thought to have a role in the development and progression of atherosclerosis. there is developing an enthusiasm for AGEs as a potential remedial target. AGES mainly induce arterial damage and exacerbate the development of atherosclerotic plaques by triggering cell receptor-dependent signalling. The interplay of AGEs with RAGE, a transmembrane signalling receptor present across all cells important to atherosclerosis, changes cell activity, boosts expression of genes, and increases the outflow of inflammatory compounds, resulting in arterial wall injury and plaque formation. Here in this review, function of AGEs in the genesis, progression, and instability of atherosclerosis is discussed. In endothelial and smooth muscle cells, as well as platelets, the interaction of AGEs with their transmembrane cell receptor, RAGE, triggers intracellular signalling, resulting in endothelial damage, vascular smooth muscle cell function modification, and changed platelet activity.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, 844102, Dist:Vaishali, Hajipur, Bihar, India.
| | - Boddu Veerabadra Siva
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, 844102, Dist:Vaishali, Hajipur, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, 844102, Dist:Vaishali, Hajipur, Bihar, India
| |
Collapse
|
19
|
Steenbeke M, Speeckaert R, Desmedt S, Glorieux G, Delanghe JR, Speeckaert MM. The Role of Advanced Glycation End Products and Its Soluble Receptor in Kidney Diseases. Int J Mol Sci 2022; 23:ijms23073439. [PMID: 35408796 PMCID: PMC8998875 DOI: 10.3390/ijms23073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are more prone to oxidative stress and chronic inflammation, which may lead to an increase in the synthesis of advanced glycation end products (AGEs). Because AGEs are mostly removed by healthy kidneys, AGE accumulation is a result of both increased production and decreased kidney clearance. On the other hand, AGEs may potentially hasten decreasing kidney function in CKD patients, and are independently related to all-cause mortality. They are one of the non-traditional risk factors that play a significant role in the underlying processes that lead to excessive cardiovascular disease in CKD patients. When AGEs interact with their cell-bound receptor (RAGE), cell dysfunction is initiated by activating nuclear factor kappa-B (NF-κB), increasing the production and release of inflammatory cytokines. Alterations in the AGE-RAGE system have been related to the development of several chronic kidney diseases. Soluble RAGE (sRAGE) is a decoy receptor that suppresses membrane-bound RAGE activation and AGE-RAGE-related toxicity. sRAGE, and more specifically, the AGE/sRAGE ratio, may be promising tools for predicting the prognosis of kidney diseases. In the present review, we discuss the potential role of AGEs and sRAGE as biomarkers in different kidney pathologies.
Collapse
Affiliation(s)
- Mieke Steenbeke
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000 Ghent, Belgium; (M.S.); (S.D.); (G.G.)
| | - Reinhart Speeckaert
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium;
- Research Foundation Flanders, 1000 Brussels, Belgium
| | - Stéphanie Desmedt
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000 Ghent, Belgium; (M.S.); (S.D.); (G.G.)
| | - Griet Glorieux
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000 Ghent, Belgium; (M.S.); (S.D.); (G.G.)
| | - Joris R. Delanghe
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Marijn M. Speeckaert
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000 Ghent, Belgium; (M.S.); (S.D.); (G.G.)
- Research Foundation Flanders, 1000 Brussels, Belgium
- Correspondence:
| |
Collapse
|
20
|
Koska J, Gerstein HC, Beisswenger PJ, Reaven PD. Advanced Glycation End Products Predict Loss of Renal Function and High-Risk Chronic Kidney Disease in Type 2 Diabetes. Diabetes Care 2022; 45:684-691. [PMID: 35051276 PMCID: PMC8918197 DOI: 10.2337/dc21-2196] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/18/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To evaluate the association of a multicomponent advanced glycation end product (AGE) panel with decline in kidney function and its utility in predicting renal function loss (RFL) when added to routine clinical measures in type 2 diabetes. RESEARCH DESIGN AND METHODS Carboxymethyl and carboxyethyl lysine and methylglyoxal, 3-deoxyglucosone, and glyoxal hydroimidazolones were measured in baseline serum and plasma samples, respectively, from Action to Control Cardiovascular Risk in Diabetes (ACCORD) (n = 1,150) and Veterans Affairs Diabetes Trial (VADT) (n = 447) participants. A composite AGE score was calculated from individual AGE z scores. The primary outcome was a sustained 30% decline in estimated glomerular filtration rate (eGFR) (30% RFL in both cohorts). Secondary outcomes (in ACCORD) were 40% RFL, macroalbuminuria, and high-risk chronic kidney disease (hrCKD). RESULTS After adjustment for baseline and follow-up HbA1c and other risk factors in ACCORD, the AGE score was associated with reduction in eGFR (β-estimate -0.66 mL/min ⋅ 1.73 m2 per year; P = 0.001), 30% RFL (hazard ratio 1.42 [95% CI 1.13-1.78]; P = 0.003), 40% RFL (1.40 [1.13-1.74]; P = 0.003), macroalbuminuria (1.53 [1.13-2.06]; P = 0.006), and hrCKD (1.88 [1.37-2.57]; P < 0.0001). AGE score improved net reclassification (NRI) and relative integrated discrimination (IDI) for 30% RFL (NRI 23%; P = 0.02) (relative IDI 7%; P = 0.009). In VADT, the AGE score calculated by the ACCORD-derived coefficients was associated with 30% RFL (1.37 [1.03-1.82); P = 0.03) and improved NRI (24%; P = 0.03) but not IDI (P = 0.18). CONCLUSIONS These data provide further support for a causal role of AGEs in diabetic nephropathy independently of glycemic control and suggest utility of the composite AGE panel in predicting long-term decline in renal function.
Collapse
Affiliation(s)
- Juraj Koska
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ
| | | | | | - Peter D Reaven
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ.,University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| |
Collapse
|
21
|
Protective role of activating PPARγ in advanced glycation end products-induced impairment of coronary artery vasodilation via inhibiting p38 phosphorylation and reactive oxygen species production. Biomed Pharmacother 2022; 147:112641. [DOI: 10.1016/j.biopha.2022.112641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 12/23/2022] Open
|
22
|
Wang M, Li Y, Li S, Lv J. Endothelial Dysfunction and Diabetic Cardiomyopathy. Front Endocrinol (Lausanne) 2022; 13:851941. [PMID: 35464057 PMCID: PMC9021409 DOI: 10.3389/fendo.2022.851941] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/14/2022] [Indexed: 12/22/2022] Open
Abstract
The cardiovascular complications contribute to a majority of diabetes associated morbidity and mortality, accounting for 44% of death in those patients with type 1 diabetes mellitus (DM) and 52% of deaths in type 2 DM. Diabetes elicits cardiovascular dysfunction through 2 major mechanisms: ischemic and non-ischemic. Non-ischemic injury is usually under-recognized although common in DM patients, and also a pathogenic factor of heart failure in those diabetic individuals complicated with ischemic heart disease. Diabetic cardiomyopathy (DCM) is defined as a heart disease in which the myocardium is structurally and functionally abnormal in the absence of coronary artery disease, hypertensive, valvular, or congenital heart disorders in diabetic patients, theoretically caused by non-ischemic injury solely. Current therapeutic strategies targeting DCM mainly address the increased blood glucose levels, however, the effects on heart function are disappointed. Accumulating data indicate endothelial dysfunction plays a critical role in the initiation and development of DCM. Hyperglycemia, hyperinsulinemia, and insulin resistance cause the damages of endothelial function, including barrier dysfunction, impaired nitric oxide (NO) activity, excessive reactive oxygen species (ROS) production, oxidative stress, and inflammatory dysregulation. In turn, endothelial dysfunction promotes impaired myocardial metabolism, intracellular Ca2+ mishandling, endoplasmic reticulum (ER) stress, mitochondrial defect, accumulation of advanced glycation end products, and extracellular matrix (ECM) deposit, leads to cardiac stiffness, fibrosis, and remodeling, eventually results in cardiac diastolic dysfunction, systolic dysfunction, and heart failure. While endothelial dysfunction is closely related to cardiac dysfunction and heart failure seen in DCM, clinical strategies for restoring endothelial function are still missing. This review summarizes the timely findings related to the effects of endothelial dysfunction on the disorder of myocardium as well as cardiac function, provides mechanical insights in pathogenesis and pathophysiology of DCM developing, and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Moran Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongsheng Li
- Department of Emergency, Tongji Hospital, Tongji Medical College, Science and Technology, Huazhong University, Wuhan, China
- *Correspondence: Yongsheng Li, ; Sheng Li, ;
| | - Sheng Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yongsheng Li, ; Sheng Li, ;
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Xiao ZL, Ma LP, Yang DF, Yang M, Li ZY, Chen MF. Profilin-1 is involved in macroangiopathy induced by advanced glycation end products via vascular remodeling and inflammation. World J Diabetes 2021; 12:1875-1893. [PMID: 34888013 PMCID: PMC8613658 DOI: 10.4239/wjd.v12.i11.1875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/29/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The accumulation of advanced glycation end products (AGEs) have been implicated in the development and progression of diabetic vasculopathy. However, the role of profilin-1 as a multifunctional actin-binding protein in AGEs-induced atherosclerosis (AS) is largely unknown.
AIM To explore the potential role of profilin-1 in the pathogenesis of AS induced by AGEs, particularly in relation to the Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) signaling pathway.
METHODS Eighty-nine individuals undergoing coronary angiography were enrolled in the study. Plasma cytokine levels were detected using ELISA kits. Rat aortic vascular smooth muscle cells (RASMCs) were incubated with different compounds for different times. Cell proliferation was determined by performing the MTT assay and EdU staining. An AGEs-induced vascular remodeling model was established in rats and histological and immunohistochemical analyses were performed. The mRNA and protein levels were detected using real-time PCR and Western blot analysis, respectively. In vivo, shRNA transfection was performed to verify the role of profilin-1 in AGEs-induced proatherogenic mediator release and aortic remodeling. Statistical analyses were performed using SPSS 22.0 software.
RESULTS Compared with the control group, plasma levels of profilin-1 and receptor for AGEs (RAGE) were significantly increased in patients with coronary artery disease, especially in those complicated with diabetes mellitus (P < 0.01). The levels of profilin-1 were positively correlated with the levels of RAGE (P < 0.01); additionally, the levels of both molecules were positively associated with the degree of coronary artery stenosis (P < 0.01). In vivo, tail vein injections of AGEs induced the release of proatherogenic mediators, such as asymmetric dimethylarginine, intercellular adhesion molecule-1, and the N-terminus of procollagen III peptide, concomitant with apparent aortic morphological changes and significantly upregulated expression of the profilin-1 mRNA and protein in the thoracic aorta (P < 0.05 or P < 0.01). Downregulation of profilin-1 expression with an shRNA significantly attenuated AGEs-induced proatherogenic mediator release (P < 0.05) and aortic remodeling. In vitro, incubation of vascular smooth muscle cells (VSMCs) with AGEs significantly promoted cell proliferation and upregulated the expression of the profilin-1 mRNA and protein (P < 0.05). AGEs (200 μg/mL, 24 h) significantly upregulated the expression of the STAT3 mRNA and protein and JAK2 protein, which was blocked by a JAK2 inhibitor (T3042-1) and/or STAT3 inhibitor (T6308-1) (P < 0.05). In addition, pretreatment with T3042-1 or T6308-1 significantly inhibited AGEs-induced RASMC proliferation (P < 0.05).
CONCLUSION AGEs induce proatherogenic events such as VSMC proliferation, proatherogenic mediator release, and vascular remodeling, changes that can be attenuated by silencing profilin-1 expression. These results suggest a crucial role for profilin-1 in AGEs-induced vasculopathy.
Collapse
Affiliation(s)
- Zhi-Lin Xiao
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Li-Ping Ma
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong Province, China
| | - Da-Feng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Mei Yang
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Zhen-Yu Li
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Mei-Fang Chen
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
24
|
Hansen L, Joseph G, Valdivia A, Taylor WR. Satellite Cell Expression of RAGE (Receptor for Advanced Glycation end Products) Is Important for Collateral Vessel Formation. J Am Heart Assoc 2021; 10:e022127. [PMID: 34689598 PMCID: PMC8751830 DOI: 10.1161/jaha.120.022127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The growth and remodeling of vascular networks is an important component of the prognosis for patients with peripheral artery disease. One protein that has been previously implicated to play a role in this process is RAGE (receptor for advanced glycation end products). This study sought to determine the cellular source of RAGE in the ischemic hind limb and the role of RAGE signaling in this cell type. Methods and Results Using a hind limb ischemia model of vascular growth, this study found skeletal muscle satellite cells to be a novel major cellular source of RAGE in ischemic tissue by both staining and cellular sorting. Although wild-type satellite cells increased tumor necrosis factor-α and monocyte chemoattractant protein-1 production in response to ischemia in vivo and a RAGE ligand in vitro, satellite cells from RAGE knockout mice lacked the increase in cytokine production both in vivo in response to ischemia and in vitro after stimuli with the RAGE ligand high-mobility group box 1. Furthermore, encapsulated wild-type satellite cells improved perfusion after hind limb ischemia surgery by both perfusion staining and vessel quantification, but RAGE knockout satellite cells provided no improvement over empty capsules. Conclusions Thus, RAGE expression and signaling in satellite cells is crucial for their response to stimuli and angiogenic and arteriogenic functions.
Collapse
Affiliation(s)
- Laura Hansen
- Division of Cardiology Department of Medicine Emory University Atlanta GA.,Division of Cardiology Atlanta Veterans Affairs Medical Center Decatur GA
| | - Giji Joseph
- Division of Cardiology Department of Medicine Emory University Atlanta GA
| | - Alejandra Valdivia
- Division of Cardiology Department of Medicine Emory University Atlanta GA
| | - W Robert Taylor
- Division of Cardiology Department of Medicine Emory University Atlanta GA.,Division of Cardiology Atlanta Veterans Affairs Medical Center Decatur GA.,The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA
| |
Collapse
|
25
|
Solly EL, Psaltis PJ, Bursill CA, Tan JTM. The Role of miR-181c in Mechanisms of Diabetes-Impaired Angiogenesis: An Emerging Therapeutic Target for Diabetic Vascular Complications. Front Pharmacol 2021; 12:718679. [PMID: 34483928 PMCID: PMC8414254 DOI: 10.3389/fphar.2021.718679] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is estimated to affect up to 700 million people by the year 2045, contributing to an immense health and economic burden. People living with diabetes have a higher risk of developing numerous debilitating vascular complications, leading to an increased need for medical care, a reduced quality of life and increased risk of early death. Current treatments are not satisfactory for many patients who suffer from impaired angiogenesis in response to ischaemia, increasing their risk of ischaemic cardiovascular conditions. These vascular pathologies are characterised by endothelial dysfunction and abnormal angiogenesis, amongst a host of impaired signaling pathways. Therapeutic stimulation of angiogenesis holds promise for the treatment of diabetic vascular complications that stem from impaired ischaemic responses. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis to improve ischaemic complications such as ischaemic heart disease and peripheral artery disease, highlighting the immense unmet need. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis in a clinical setting, highlighting the immense unmet need. MicroRNAs (miRNAs) are emerging as powerful targets for multifaceted diseases including diabetes and cardiovascular disease. This review highlights the potential role of microRNAs as therapeutic targets for rescuing diabetes-impaired angiogenesis, with a specific focus on miR-181c, which we have previously identified as an important angiogenic regulator. Here we summarise the pathways currently known to be regulated by miR-181c, which include the classical angiogenesis pathways that are dysregulated in diabetes, mitochondrial function and axonal guidance, and describe how these relate both directly and indirectly to angiogenesis. The pleiotropic actions of miR-181c across multiple key angiogenic signaling pathways and critical cellular processes highlight its therapeutic potential as a novel target for treating diabetic vascular complications.
Collapse
Affiliation(s)
- Emma L Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
26
|
Bora S, Shankarrao Adole P. Carbonyl stress in diabetics with acute coronary syndrome. Clin Chim Acta 2021; 520:78-86. [PMID: 34090879 DOI: 10.1016/j.cca.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 01/17/2023]
Abstract
The prevalence and incidence of diabetes mellitus (DM) are increasing worldwide bringing with it a significantly higher rate of complications. Various mechanisms such as carbonyl stress, polyol pathway, oxidative stress, hexosamine pathways, diacylglycerol/protein kinase-C activation, etc., are responsible for the pathogenesis of DM and its complications. Persistent hyperglycaemia and inhibition of metabolising and detoxifying enzymes lead to the excessive synthesis of carbonyl compounds such as methylglyoxal, glyoxal, and 3-deoxyglucosone, resulting in carbonyl stress. The substrates, metabolizing and detoxifying enzymes of carbonyl compounds are discussed. The mechanistic roles of carbonyl compounds and advanced glycation end products (AGEs) in atherosclerosis, insulin resistance, thrombogenicity, and endothelial dysfunction in animal and cell culture model of DM and patients with DM are summarised. Because of the essential role of carbonyl stress, therapeutics are aimed at scavenging, metabolizing, detoxifying, and inhibiting carbonyl compounds or AGEs so that their harmful effects are minimized. Clinically used drugs, plants extracts and miscellaneous chemical with antiglycation properties are used in an animal model of DM to alleviates the impact of carbonyl compounds. Extensive clinical trials with derivatisation of available antiglycation agents to increase the bioavailability and decrease side effects are warranted further.
Collapse
Affiliation(s)
- Sushmita Bora
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605 006, India
| | - Prashant Shankarrao Adole
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605 006, India.
| |
Collapse
|
27
|
Apolipoprotein E Gene Polymorphism, Glycated Hemoglobin, and Peripheral Arterial Disease Risk in Chinese Type 2 Diabetic Patients. DISEASE MARKERS 2021; 2020:6040525. [PMID: 32211081 PMCID: PMC7085830 DOI: 10.1155/2020/6040525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 11/17/2022]
Abstract
Background The apolipoprotein E (ApoE) gene polymorphism has been found to influence plasma lipid concentration, and its correlation with peripheral arterial disease (PAD) has been investigated. However, it is unclear whether ApoE is associated with PAD in Chinese type 2 diabetes mellitus (T2DM) patients. Therefore, our study is aimed at investigating the relationship between the ApoE gene polymorphism and PAD in Chinese T2DM patients. Methods A total of 192 T2DM patients were divided into two groups: T2DM and T2DM with PAD. The clinical and biochemical parameters were obtained. Polymerase chain reaction was used to identify the genotypes of ApoE. The multivariable logistic regression analysis was used to identify the possible risk factor for PAD. Results There were no significant differences in the genotype and allele frequencies of ApoE between the T2DM and T2DM with PAD groups. However, the T2DM with PAD group tended to have more ε4/ε4/ Conclusions These results demonstrated that there was no evidence of a relationship between ApoE and PAD.
Collapse
|
28
|
Vitamin D Deficiency May Not Be an Independent Risk Factor for Peripheral Arterial Disease in Middle-Aged and Elderly Patients with Type 2 Diabetes in China. DISEASE MARKERS 2020; 2020:8854717. [PMID: 33299499 PMCID: PMC7710426 DOI: 10.1155/2020/8854717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022]
Abstract
Background Vitamin D deficiency can lead to the increased severity and prevalence of metabolic disorders. However, the relationship between levels of 25-hydroxyvitamin D (25(OH)D) and peripheral arterial disease (PAD) is controversial. Therefore, the purpose of our study was to explore the relationship between 25(OH)D levels and PAD in middle-aged and elderly type 2 diabetes mellitus (T2DM) patients in China. Methods In this study, a total of 183 patients with T2DM were enrolled and categorized into groups with or without PAD. Clinical and biochemical parameters were assessed, and a Pearson analysis was used to identify a possible association between levels of 25(OH)D and glycated hemoglobin (HbA1c). Some biochemical parameters were also assessed in the T2DM patients with PAD according to vitamin D status. Interactions were also explored among HbA1c control, 25(OH)D levels, and PAD. The possible risk factors for PAD were measured by multivariable logistic regression analyses. Results Firstly, the parameters including age, HbA1c, and disease duration between T2DM and T2DM+PAD groups showed significantly different. In addition, the frequency of smoking in the group of T2DM patients was significantly less than that in the T2DM patients with the PAD group, while the frequency of well-controlled HbA1c in the patients with T2DM was significantly higher. There is a trend that the levels of 25(OH)D and HbA1c are correlated, but no interactions among vitamin D deficiency, HbA1c control, and PAD were found. However, HbA1c significantly differed between groups with vitamin D deficiency and insufficiency in the T2DM patients with PAD. According to the multivariate logistic regression analyses, the PAD risk factors of T2DM patients were family history of diabetes, smoking, age, disease duration, HbA1c, and LDL. Conclusions The findings demonstrate that the deficiency of vitamin D level is not related to PAD, but HbA1c may be linked to the presence of PAD in middle-aged and elderly patients with T2DM in China.
Collapse
|
29
|
Saz-Lara A, Álvarez-Bueno C, Martínez-Vizcaíno V, Notario-Pacheco B, Sequí-Dominguez I, Cavero-Redondo I. Are Advanced Glycation End Products in Skin Associated with Vascular Dysfunction Markers? A Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17186936. [PMID: 32972023 PMCID: PMC7559442 DOI: 10.3390/ijerph17186936] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 12/22/2022]
Abstract
Evidence exists regarding the association between advanced glycation end products and different cardiovascular disease subclinical processes, such as arterial stiffness and atherosclerosis. With this systematic review and meta-analysis, we aimed to provide a synthesis of the evidence regarding the association of arterial stiffness measured by pulse wave velocity and atherosclerosis measured by carotid intima media thickness with skin autofluorescence. A systematic search was performed using: MEDLINE (PubMed), SCOPUS, and Web of Science, until 30 March 2020. Cross-sectional studies or baseline data from prospective longitudinal studies were considered. The DerSimonian and Laird method was used to calculate the pooled estimates of correlation coefficients and the corresponding 95% confidence intervals (CI) for the association of pulse wave velocity and carotid intima media thickness with skin autofluorescence. Twenty-five studies were included in the systematic review and meta-analysis, including 6306 subjects. The pooled correlation coefficient was 0.25 (95% CI: 0.18, 0.31) for pulse wave velocity and skin autofluorescence, and 0.31 (95% CI: 0.25, 0.38) for carotid intima media thickness and skin autofluorescence. This systematic review and meta-analysis provide a synthesis of the evidence showing a positive weak association of pulse wave velocity and carotid intima media thickness with skin autofluorescence.
Collapse
Affiliation(s)
- Alicia Saz-Lara
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16171 Cuenca, Spain; (A.S.-L.); (V.M.-V.); (B.N.-P.); (I.S.-D.); (I.C.-R.)
| | - Celia Álvarez-Bueno
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16171 Cuenca, Spain; (A.S.-L.); (V.M.-V.); (B.N.-P.); (I.S.-D.); (I.C.-R.)
- Universidad Politécnica y Artística del Paraguay, 001518 Asuncion, Paraguay
- Correspondence:
| | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16171 Cuenca, Spain; (A.S.-L.); (V.M.-V.); (B.N.-P.); (I.S.-D.); (I.C.-R.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 3460000 Talca, Chile
| | - Blanca Notario-Pacheco
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16171 Cuenca, Spain; (A.S.-L.); (V.M.-V.); (B.N.-P.); (I.S.-D.); (I.C.-R.)
| | - Irene Sequí-Dominguez
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16171 Cuenca, Spain; (A.S.-L.); (V.M.-V.); (B.N.-P.); (I.S.-D.); (I.C.-R.)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16171 Cuenca, Spain; (A.S.-L.); (V.M.-V.); (B.N.-P.); (I.S.-D.); (I.C.-R.)
- Universidad Politécnica y Artística del Paraguay, 001518 Asuncion, Paraguay
| |
Collapse
|
30
|
Yubero-Serrano EM, Pérez-Martínez P. Advanced Glycation End Products and Their Involvement in Cardiovascular Disease. Angiology 2020; 71:698-700. [PMID: 32242451 DOI: 10.1177/0003319720916301] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Elena M Yubero-Serrano
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba, Reina Sofia University Hospital, University of Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Pablo Pérez-Martínez
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba, Reina Sofia University Hospital, University of Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Haesen S, Cöl Ü, Schurgers W, Evens L, Verboven M, Driesen RB, Bronckaers A, Lambrichts I, Deluyker D, Bito V. Glycolaldehyde-modified proteins cause adverse functional and structural aortic remodeling leading to cardiac pressure overload. Sci Rep 2020; 10:12220. [PMID: 32699285 PMCID: PMC7376068 DOI: 10.1038/s41598-020-68974-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the role of advanced glycation end products (AGEs) in the development of diabetic vascular complications and cardiovascular diseases (CVDs). We have shown that high-molecular-weight AGEs (HMW-AGEs), present in our Western diet, impair cardiac function. Whether HMW-AGEs affect vascular function remains unknown. In this study, we aimed to investigate the impact of chronic HMW-AGEs exposure on vascular function and structure. Adult male Sprague Dawley rats were daily injected with HMW-AGEs or control solution for 6 weeks. HMW-AGEs animals showed intracardiac pressure overload, characterized by increased systolic and mean pressures. The contraction response to PE was increased in aortic rings from the HMW-AGEs group. Relaxation in response to ACh, but not SNP, was impaired by HMW-AGEs. This was associated with reduced plasma cyclic GMP levels. SOD restored ACh-induced relaxation of HMW-AGEs animals to control levels, accompanied by a reduced half-maximal effective dose (EC50). Finally, collagen deposition and intima-media thickness of the aortic vessel wall were increased with HMW-AGEs. Our data demonstrate that chronic HMW-AGEs exposure causes adverse vascular remodelling. This is characterised by disturbed vasomotor function due to increased oxidative stress and structural changes in the aorta, suggesting an important contribution of HMW-AGEs in the development of CVDs.
Collapse
Affiliation(s)
- Sibren Haesen
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Ümare Cöl
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Wouter Schurgers
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Lize Evens
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Maxim Verboven
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Ronald B Driesen
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Ivo Lambrichts
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Dorien Deluyker
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Virginie Bito
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium.
| |
Collapse
|
32
|
Li X, Tao Y, Wang X, Wang T, Liu J. Advanced glycosylation end products (AGEs) controls proliferation, invasion and permeability through orchestrating ARHGAP18/RhoA pathway in human umbilical vein endothelial cells. Glycoconj J 2020; 37:209-219. [PMID: 32016689 DOI: 10.1007/s10719-020-09908-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022]
Abstract
Diabetic vascular complications caused by endothelial dysfunction play an important role in the pathogenesis of diabetic foot. A well understanding of the role of endothelial dysfunction in diabetic foot vasculopathy will help to further reveal the pathogenesis of diabetic foot. This study aimed to assess whether the RhoA/ROCK signaling pathway is controlled by Rho GTPase-activating proteins (RhoGAP, ARHGAP) and advanced glycosylation end products (AGEs), and to clarify the roles of ARHGAP and AGEs in the RhoA/ROCK signaling pathway or the mechanism by which AGEs regulated RhoA. Real-time PCR was applied to detect gene expression. Manipulation of endothelial biological functions by ARHGAP18 and AGEs were studied via cell counting kit-8 (CCK-8), Western blot, transwell, FITC-Dextran and TEER permeability experiments. RhoA-specific inhibitor Y-27632 was used to silence the activity of RhoA. Dual Luciferase Reporter Assay, Western blot and ELISA assays were used to detect molecular mechanism of endothelial biological functions. In this study, we found that ARHGAP18 was negatively correlated with RhoA, and the expression of ARHGAP18 in human umbilical vein endothelial cells (HUVECs) was decreased with gradient-increased AGEs. Furthermore, AGEs and ARHGAP18 could orchestrate RhoA activity, then activate NF-κB signaling pathway, affect the structural and morphological of VE-cadherin and tight junction protein, and cause endothelial cell contraction, thereby increasing permeability of endothelial cells. In conclusion, AGEs and ARHGAP18 orchestrate cell proliferation, invasion and permeability by controlling the RhoA/ROCK signaling pathway, affecting NF-κB signaling pathway as well as the structure and morphology of VE-cadherin and tight junction protein, and regulating endothelial cell contraction.
Collapse
Affiliation(s)
- Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Yue Tao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Xiaojun Wang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Tao Wang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Jianjun Liu
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China.
| |
Collapse
|
33
|
Endothelial Dysfunction and Advanced Glycation End Products in Patients with Newly Diagnosed Versus Established Diabetes: From the CORDIOPREV Study. Nutrients 2020; 12:nu12010238. [PMID: 31963378 PMCID: PMC7019746 DOI: 10.3390/nu12010238] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 01/22/2023] Open
Abstract
Endothelial dysfunction and intima-media thickness of common carotid arteries (IMT-CC) are considered subclinical markers of atherosclerotic cardiovascular disease (ASCVD). Advanced glycation end products (AGEs) are increased in type 2 diabetes mellitus (T2DM) patients, compared with non-diabetics, being implicated in micro- and macrovascular complications. Our aim was to compare serum AGEs levels and subclinical atherosclerotic markers between patients with established and newly diagnosed T2DM. Among 540 patients with T2DM and coronary heart disease from the CORDIOPREV study, 350 patients had established T2DM and 190 patients had newly diagnosed T2DM. Serum levels of AGEs (methylglyoxal (MG) and N-carboxymethyl lysine (CML)) and subclinical atherosclerotic markers (brachial flow-mediated vasodilation (FMD) and IMT-CC) were measured. AGEs levels (all p < 0.001) and IMT-CC (p = 0.025) were higher in patients with established vs. newly diagnosed T2DM, whereas FMD did not differ between the two groups. Patients with established T2DM and severe endothelial dysfunction (i.e., FMD < 2%) had higher serum MG levels, IMT-CC, HOMA-IR and fasting insulin levels than those with newly diagnosed T2DM and non-severe endothelial dysfunction (i.e., FMD ≥ 2%) (all p < 0.05). Serum CML levels were greater in patients with established vs. newly diagnosed T2DM, regardless of endothelial dysfunction severity. Serum AGEs levels and IMT-CC were significantly higher in patients with established vs. newly diagnosed T2DM, highlighting the progressively increased risk of ASCVD in the course of T2DM. Establishing therapeutic strategies to reduce AGEs production and delay the onset of cardiovascular complications in newly diagnosed T2DM patients or minimize ASCVD risk in established T2DM patients is needed.
Collapse
|
34
|
Siasos G, Skotsimara G, Oikonomou E, Sagris M, Vasiliki-Chara M, Bletsa E, Stampouloglou P, Theofilis P, Charalampous G, Tousoulis D. Antithrombotic Treatment in Diabetes Mellitus: A Review of the Literature about Antiplatelet and Anticoagulation Strategies Used for Diabetic Patients in Primary and Secondary Prevention. Curr Pharm Des 2020; 26:2780-2788. [PMID: 32303164 DOI: 10.2174/1381612826666200417145605] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Diabetes mellitus (DM) is on the rise globally. Its prevalence has nearly doubled during the last two decades and it is estimated to affect 8.8% of the global population. Cardiovascular disease (CVD) is the leading cause of death in the diabetic population and despite modern anti-inflammatory and cardioprotective therapeutic strategies, diabetic patients have at least a twice fold risk of cardiovascular events. The prothrombotic state in DM is associated with multiple determinants such as platelet alterations, oxidative stress, endothelial changes, circulating mediators. Thus, proper antithrombotic strategies to reduce the risk of CVD in this population are critical. METHODS This article reviews the current antiplatelet and anticoagulant agents in the aspect of primary and secondary prevention of CVD in the diabetic population. RESULTS The use of aspirin may be considered only at high-risk patients in the absence of contraindications. Cangrelor was not inferior to clopidogrel in preventing the composite outcome of CV death, myocardial infarction and revascularization without increasing major bleeding. Triple therapy in the subpopulation with DM significantly reduced the composite primary outcome of CV death, myocardial infarction or repeat target lesion revascularization. That was not the case for stent thrombosis, which was similar in both groups. Importantly, triple therapy did not result in increased bleeding complications, which were similar in both groups. However, cilostazol is linked to various adverse effects (e.g., headache, palpitations, and gastrointestinal disturbances) that drive many patients to withdrawal. CONCLUSION In conclusion, DM is a rapidly growing disease that increases the risk of CVD, AF, and CV mortality. Proper antithrombotic strategies to reduce CVD risk in DM are a necessity. Moreover, new antithrombotic treatments and combination therapies may play a critical role to overcome antiplatelet resistance in DM patients and reduce morbidity and mortality attributed to CVD.
Collapse
Affiliation(s)
- Gerasimos Siasos
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Georgia Skotsimara
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Evangelos Oikonomou
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Marios Sagris
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Mystakidi Vasiliki-Chara
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Evanthia Bletsa
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Panagiota Stampouloglou
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Panagiotis Theofilis
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Georgios Charalampous
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| | - Dimitris Tousoulis
- First Department of Cardiology, Hippokration Hospital, Athens Medical School, 114 Vas. Sofias avenue, 11527, Athens, Greece
| |
Collapse
|
35
|
Amplification of the COX/TXS/TP receptor pathway enhances uridine diphosphate-induced contraction by advanced glycation end products in rat carotid arteries. Pflugers Arch 2019; 471:1505-1517. [PMID: 31736003 DOI: 10.1007/s00424-019-02330-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022]
Abstract
Advanced glycation end products (AGEs) play a pivotal role in vascular functions under various pathophysiological conditions. Although uridine diphosphate (UDP) is an important extracellular nucleotide, the relationship between AGEs and UDP regarding their effect on vascular functions remains unclear. Therefore, we investigated the effects of AGE-bovine serum albumin (AGE-BSA) on UDP-mediated responses in rat thoracic aorta and carotid arteries. In rat thoracic aorta, UDP-induced relaxation was observed and this relaxation was similar between control (1.0 v/v% PBS) and AGE-BSA-treated (0.1 mg/mL for 60 min) groups. In contrast, contraction but not relaxation was obtained following UDP application to carotid arteries with and without endothelia; contraction was greater in the AGE-BSA-treated group than in the control group. The difference in UDP-induced contraction between the two groups was not abolished by the use of a nitric oxide synthase (NOS) inhibitor, whereas it was abolished by the use of cyclooxygenase (COX), thromboxane synthase (TXS), and thromboxane-prostanoid (TP) receptor antagonist. Further, the difference in UDP-induced contraction was not abolished by the use of a cPLA2 inhibitor, whereas it was abolished by the use of an iPLA2 inhibitor. UDP increased TXA2 release in both groups, and its level was similar in both groups. Moreover, the release of PGE2, PGF2α, and PGI2 was similar among the groups. Under NOS inhibition, TP receptor agonist-induced contraction increased in the AGE-BSA-treated group (vs. control group). In conclusion, the increase in UDP-induced carotid arterial contraction by AGE-BSA can be attributed to an increase in the COX/TXS/TP receptor pathway, particularly, TP receptor signaling.
Collapse
|
36
|
Wang CC, Shen MY, Chang KC, Wang GJ, Liu SH, Chang CT. Skin autofluorescence is associated with rapid renal function decline in subjects at increased risk of coronary artery disease. PLoS One 2019; 14:e0217203. [PMID: 31116778 PMCID: PMC6530849 DOI: 10.1371/journal.pone.0217203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 05/08/2019] [Indexed: 12/24/2022] Open
Abstract
Skin autofluorescence (AF) has been validated as a tool for estimating tissue advanced glycation end products (AGEs) accumulation and predicting long-term cardiovascular outcomes. However, whether measurements of skin AF could predict renal function decline remains controversial. From April, 2014 to April, 2015, we enrolled 245 subjects with at least two conventional risk factors for coronary artery disease (CAD). All were measured for body height and weight, blood pressure, plasma creatinine level, and skin AF at the start of the study. Baseline demographics and laboratory tests data were obtained by chart reviews and patient interviews. Serial plasma creatinine levels were followed regularly every 6-12 months for 2 years. In a stepwise multivariate linear regression analysis, skin AF, was an independent factor for predicting the relative renal function decline rate after adjustment of multiple covariates (ß = -0.036±0.016; p = 0.03). Subgroups analysis revealed that skin AF was a significant factor for relative renal function decline rate in subgroups of age < 65 years (ß = -0.068±0.024; p = 0.02), male sex (ß = -0.053±0.016; p< 0.01), body mass index≧25 Kg/m2(ß = -0.042±0.021; p = 0.04), and estimated glomerular filtration rate ≥ 60 ml/min/1.73m2(ß = -0.043±0.020; p = 0.04). However, only an interaction between skin AF and age attained significance (p for interaction = 0.04). Skin AF is a useful predictor for renal function decline in patients at increased risk of CAD.
Collapse
Affiliation(s)
- Chun-Cheng Wang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,Cardiovascular Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Yi Shen
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Cardiovascular Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Kuan-Cheng Chang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,Cardiovascular Research Laboratory, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Guei-Jane Wang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Shu-Hui Liu
- Cardiovascular Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Chiz-Tzung Chang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Cardiovascular Research Laboratory, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
37
|
Nawaz IM, Rezzola S, Cancarini A, Russo A, Costagliola C, Semeraro F, Presta M. Human vitreous in proliferative diabetic retinopathy: Characterization and translational implications. Prog Retin Eye Res 2019; 72:100756. [PMID: 30951889 DOI: 10.1016/j.preteyeres.2019.03.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
Abstract
Diabetic retinopathy (DR) is one of the leading causes of visual impairment in the working-age population. DR is a progressive eye disease caused by long-term accumulation of hyperglycaemia-mediated pathological alterations in the retina of diabetic patients. DR begins with asymptomatic retinal abnormalities and may progress to advanced-stage proliferative diabetic retinopathy (PDR), characterized by neovascularization or preretinal/vitreous haemorrhages. The vitreous, a transparent gel that fills the posterior cavity of the eye, plays a vital role in maintaining ocular function. Structural and molecular alterations of the vitreous, observed during DR progression, are consequences of metabolic and functional modifications of the retinal tissue. Thus, vitreal alterations reflect the pathological events occurring at the vitreoretinal interface. These events are caused by hypoxic, oxidative, inflammatory, neurodegenerative, and leukostatic conditions that occur during diabetes. Conversely, PDR vitreous can exert pathological effects on the diabetic retina, resulting in activation of a vicious cycle that contributes to disease progression. In this review, we recapitulate the major pathological features of DR/PDR, and focus on the structural and molecular changes that characterize the vitreal structure and composition during DR and progression to PDR. In PDR, vitreous represents a reservoir of pathological signalling molecules. Therefore, in this review we discuss how studying the biological activity of the vitreous in different in vitro, ex vivo, and in vivo experimental models can provide insights into the pathogenesis of PDR. In addition, the vitreous from PDR patients can represent a novel tool to obtain preclinical experimental evidences for the development and characterization of new therapeutic drug candidates for PDR therapy.
Collapse
Affiliation(s)
- Imtiaz M Nawaz
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Anna Cancarini
- Department of Ophthalmology, University of Brescia, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Brescia, Italy
| | - Ciro Costagliola
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Italy.
| |
Collapse
|
38
|
Sun W, Gao Y, Ding Y, Cao Y, Chen J, Lv G, Lu J, Yu B, Peng M, Xu H, Sun Y. Catalpol ameliorates advanced glycation end product-induced dysfunction of glomerular endothelial cells via regulating nitric oxide synthesis by inducible nitric oxide synthase and endothelial nitric oxide synthase. IUBMB Life 2019; 71:1268-1283. [PMID: 30861639 DOI: 10.1002/iub.2032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
Abstract
Catalpol (Cat.) is an iridoid glucoside extracted from the root of Rehmannia glutinosa Libosch. In this study, we investigated whether Cat. could protect the mouse glomerular endothelial cells against the deleterious effect induced by advanced glycation end products (AGEs) and explored potential mechanisms. We found that 10 μM Cat. showed a protective effect on dead cells stimulated by AGEs. Cat. significantly decreased the expression of p-NF-κBp65 and inducible nitric oxide synthase (iNOS) and increased the expression of phosphorylated-endothelial nitric oxide synthase (p-eNOS; Ser1177), PI3K, p-Akt (Thr308), and total-Akt. Moreover, Cat. restored the integrity of glomerular endothelial barrier by increasing endothelial tight gap junction protein and ameliorated the endothelial hyperpermeability induced by AGEs via modulating the nitric oxide (NO) production. Additionally, Cat. attenuated the massive release of NO induced by AGEs, inhibiting the macrophage infiltration by modulating the NO production, accompanied by the decrease in the release of monocyte chemoattractant protein-1 and intercellular cell adhesion molecule-1 in vitro. Therefore, Cat. ameliorated AGEs-induced endothelial dysfunction via inhibiting the NF-κB/iNOS pathway and activating the PI3K/Akt/eNOS pathway. © 2019 IUBMB Life, 71(9):1268-1283, 2019.
Collapse
Affiliation(s)
- Weixiang Sun
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Yuyan Gao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Yushi Ding
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Ying Cao
- Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Jing Chen
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Gaohong Lv
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Jinfu Lu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Bin Yu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Meilin Peng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Huiqin Xu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Yun Sun
- Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| |
Collapse
|
39
|
Kosmopoulos M, Drekolias D, Zavras PD, Piperi C, Papavassiliou AG. Impact of advanced glycation end products (AGEs) signaling in coronary artery disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:611-619. [PMID: 30611860 DOI: 10.1016/j.bbadis.2019.01.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 02/08/2023]
Abstract
Coronary artery disease remains the leading cause of mortality in adult diabetic population with however, a high predominance also in non-diabetic subjects. In search of common molecular mechanisms and metabolic by-products with potential pathogenic role, increased advanced glycation end products (AGEs) present a critical biomarker for CAD development in both cases. Interaction of AGEs with their transmembrane cell receptor, RAGE in endothelial and smooth muscle cells as well as in platelets, activates intracellular signaling that leads to endothelial injury, modulation of vascular smooth muscle cell function and altered platelet activity. Furthermore, tissue accumulation of AGEs affects current treatment approaches being involved in stent restenosis. The present review provides an update of AGE-induced molecular mechanisms involved in CAD pathophysiology while it discusses emerging therapeutic interventions targeting AGE reduction and AGE-RAGE signaling with beneficial clinical outcome.
Collapse
Affiliation(s)
- Marinos Kosmopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Drekolias
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Phaedon D Zavras
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
40
|
Abstract
Circadian rhythms, meals, and exercise modulate energy metabolism. This review explores the novel hypothesis that there is an optimal time of day to exercise to improve 24 h glycemia and lipemia in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Timothy D Heden
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota - Twin Cities, Minneapolis, MN, and
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri - Columbia, Columbia, MO
| |
Collapse
|
41
|
Bansal S, Kare PK, Tripathi AK, Madhu SV. Advanced Glycation End Products: A Potential Contributor of Oxidative Stress for Cardio-Vascular Problems in Diabetes. OXIDATIVE STRESS IN HEART DISEASES 2019:437-459. [DOI: 10.1007/978-981-13-8273-4_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
42
|
Knapp M, Tu X, Wu R. Vascular endothelial dysfunction, a major mediator in diabetic cardiomyopathy. Acta Pharmacol Sin 2019; 40:1-8. [PMID: 29867137 PMCID: PMC6318313 DOI: 10.1038/s41401-018-0042-6] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022]
Abstract
Diabetes mellitus is currently a major public health problem. A common complication of diabetes is cardiac dysfunction, which is recognized as a microvascular disease that leads to morbidity and mortality in diabetic patients. While ischemic events are commonly observed in diabetic patients, the risk for developing heart failure is also increased, independent of the severity of coronary artery disease and hypertension. This diabetes-associated clinical entity is considered a distinct disease process referred to as "diabetic cardiomyopathy". However, it is not clear how diabetes promotes cardiac dysfunction. Vascular endothelial dysfunction is thought to be one of the key risk factors. The impact of diabetes on the endothelium involves several alterations, including hyperglycemia, fatty acid oxidation, reduced nitric oxide (NO), oxidative stress, inflammatory activation, and altered barrier function. The current review provides an update on mechanisms that specifically target endothelial dysfunction, which may lead to diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Maura Knapp
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Xin Tu
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Rongxue Wu
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA.
| |
Collapse
|
43
|
Yamagishi SI, Matsui T. Role of Hyperglycemia-Induced Advanced Glycation End Product (AGE) Accumulation in Atherosclerosis. Ann Vasc Dis 2018; 11:253-258. [PMID: 30402172 PMCID: PMC6200622 DOI: 10.3400/avd.ra.18-00070] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
There is a growing body of evidence that cumulative hyperglycemic exposure plays a central role in the development and progression of atherosclerotic cardiovascular disease in diabetic patients. Monosaccharides, such as glucose, fructose, and glyceraldehyde can react non-enzymatically with amino groups of proteins, lipids, nucleic acids to form senescent macromolecules termed advanced glycation end products (AGEs), whose formation and accumulation has been known to progress in diabetic patients, especially in those with a long history of disease. The sustained accumulation of AGEs could contribute to the phenomenon of metabolic memory or legacy effects observed in long-term follow-up clinical studies of diabetic patients. AGE modification alters the structural integrity and function of various types of macromolecules, and interaction of AGEs with a receptor for AGEs (RAGE) has been shown to evoke inflammatory and thrombotic reactions. Therefore, the AGE-RAGE axis is a novel therapeutic target of atherosclerotic cardiovascular disease in diabetic patients. In this paper, we briefly review the pathological role of AGEs and their receptor RAGE system in atherosclerotic cardiovascular disease, including peripheral artery disease and discuss the clinical utility of measuring AGEs in evaluating the severity of atherosclerosis in patients with diabetes.
Collapse
Affiliation(s)
- Sho-ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
44
|
Banarjee R, Sharma A, Bai S, Deshmukh A, Kulkarni M. Proteomic study of endothelial dysfunction induced by AGEs and its possible role in diabetic cardiovascular complications. J Proteomics 2018; 187:69-79. [DOI: 10.1016/j.jprot.2018.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/30/2022]
|
45
|
Khodadadi S, Zabihi NA, Niazmand S, Abbasnezhad A, Mahmoudabady M, Rezaee SA. Teucrium polium improves endothelial dysfunction by regulating eNOS and VCAM-1 genes expression and vasoreactivity in diabetic rat aorta. Biomed Pharmacother 2018; 103:1526-1530. [DOI: 10.1016/j.biopha.2018.04.158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022] Open
|
46
|
Gliemann L, Rytter N, Lindskrog M, Slingsby MHL, Åkerström T, Sylow L, Richter EA, Hellsten Y. Endothelial mechanotransduction proteins and vascular function are altered by dietary sucrose supplementation in healthy young male subjects. J Physiol 2018. [PMID: 28620941 DOI: 10.1113/jp274623] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Mechanotransduction in endothelial cells is a central mechanism in the regulation of vascular tone and vascular remodelling Mechanotransduction and vascular function may be affected by high sugar levels in plasma because of a resulting increase in oxidative stress and increased levels of advanced glycation end-products (AGE). In healthy young subjects, 2 weeks of daily supplementation with 3 × 75 g of sucrose was found to reduce blood flow in response to passive lower leg movement and in response to 12 W of knee extensor exercise. This vascular impairment was paralleled by up-regulation of platelet endothelial cell adhesion molecule (PECAM)-1, endothelial nitric oxide synthase, NADPH oxidase and Rho family GTPase Rac1 protein expression, an increased basal phosphorylation status of vascular endothelial growth factor receptor 2 and a reduced phosphorylation status of PECAM-1. There were no measurable changes in AGE levels. The findings of the present study demonstrate that daily high sucrose intake markedly affects mechanotransduction proteins and has a detrimental effect on vascular function. ABSTRACT Endothelial mechanotransduction is important for vascular function but alterations and activation of vascular mechanosensory proteins have not been investigated in humans. In endothelial cell culture, simple sugars effectively impair mechanosensor proteins. To study mechanosensor- and vascular function in humans, 12 young healthy male subjects supplemented their diet with 3 × 75 g sucrose day-1 for 14 days in a randomized cross-over design. Before and after the intervention period, the hyperaemic response to passive lower leg movement and active knee extensor exercise was determined by ultrasound doppler. A muscle biopsy was obtained from the thigh muscle before and after acute passive leg movement to allow assessment of protein amounts and the phosphorylation status of mechanosensory proteins and NADPH oxidase. The sucrose intervention led to a reduced flow response to passive movement (by 17 ± 2%) and to 12 W of active exercise (by 9 ± 1%), indicating impaired vascular function. A reduced flow response to passive and active exercise was paralleled by a significant up-regulation of platelet endothelial cell adhesion molecule (PECAM-1), endothelial nitric oxide synthase, NADPH oxidase and the Rho family GTPase Rac1 protein expression in the muscle tissue, as well as an increased basal phosphorylation status of vascular endothelial growth factor receptor 2 and a reduced phosphorylation status of PECAM-1. The phosphorylation status was not acutely altered with passive leg movement. These findings indicate that a regular intake of high levels of sucrose can impair vascular mechanotransduction and increase the oxidative stress potential, and suggest that dietary excessive sugar intake may contribute to the development of vascular disease.
Collapse
Affiliation(s)
- Lasse Gliemann
- Section for Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai Rytter
- Section for Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads Lindskrog
- Section for Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Thorbjörn Åkerström
- Section for Integrative Physiology, University of Copenhagen, Copenhagen, Denmark.,Insulin Pharmacology Department, Novo Nordisk A/S, Maaloev, Denmark
| | - Lykke Sylow
- Section Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- Section Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- Section for Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Saum K, Campos B, Celdran-Bonafonte D, Nayak L, Sangwung P, Thakar C, Roy-Chaudhury P, Owens AP. Uremic Advanced Glycation End Products and Protein-Bound Solutes Induce Endothelial Dysfunction Through Suppression of Krüppel-Like Factor 2. J Am Heart Assoc 2018; 7:e007566. [PMID: 29301761 PMCID: PMC5778969 DOI: 10.1161/jaha.117.007566] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of morbidity and mortality in patients with end-stage renal disease. The accumulation of uremic solutes in this patient population is associated with endothelial dysfunction and accelerated cardiovascular disease. In this study, we examined the impact of the uremic milieu on the endothelial transcription factor, Krüppel-like factor 2 (KLF2), a key regulator of endothelial function and activation. METHODS AND RESULTS Using serum from uremic pigs with chronic renal insufficiency, our results show that KLF2 expression is suppressed by the uremic milieu and individual uremic solutes in vitro. Specifically, KLF2 expression is significantly decreased in human umbilical vein endothelial cells after treatment with uremic porcine serum or carboxymethyllysine-modified albumin, an advanced glycation end product (AGE) known to induce endothelial dysfunction. AGE-mediated suppression of KLF2 is dependent on activation of the receptor for AGE, as measured by small interfering RNA knockdown of the receptor for AGE. Furthermore, KLF2 suppression promotes endothelial dysfunction, because adenoviral overexpression of KLF2 inhibits reactive oxygen species production and leukocyte adhesion in human umbilical vein endothelial cells. In addition, the application of hemodynamic shear stress, prolonged serum dialysis, or treatment with the receptor for AGE antagonist azeliragon (TTP488) is sufficient to prevent KLF2 suppression in vitro. To decipher the mechanism by which uremic AGEs suppress KLF2 expression, we assessed the role of the receptor for AGE in activation of nuclear factor-κB signaling, a hallmark of endothelial cell activation. Using a constitutively active form of IκBα, we show that translocation of p65 to the nucleus is necessary for KLF2 suppression after treatment with uremic AGEs. CONCLUSIONS These data identify KLF2 suppression as a consequence of the uremic milieu, which may exacerbate endothelial dysfunction and resultant cardiovascular disease.
Collapse
Affiliation(s)
- Keith Saum
- University of Cincinnati Medical Scientist Training Program, The University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Begoña Campos
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Diego Celdran-Bonafonte
- Division of Nephrology, University of Arizona College of Medicine and Banner University Medical Centers-Tucson and South and Southern Arizona Veterans Affairs Healthcare System, Tucson, AZ
| | - Lalitha Nayak
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Panjamaporn Sangwung
- Department of Physiology and Biophysics, Department of Medicine, Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Charuhas Thakar
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Prabir Roy-Chaudhury
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Nephrology, University of Arizona College of Medicine and Banner University Medical Centers-Tucson and South and Southern Arizona Veterans Affairs Healthcare System, Tucson, AZ
| | - A Phillip Owens
- Division of Cardiovascular Health and Disease, The University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
48
|
Sharma I, Tupe RS, Wallner AK, Kanwar YS. Contribution of myo-inositol oxygenase in AGE:RAGE-mediated renal tubulointerstitial injury in the context of diabetic nephropathy. Am J Physiol Renal Physiol 2017; 314:F107-F121. [PMID: 28931523 DOI: 10.1152/ajprenal.00434.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advanced glycation end products (AGEs) play a role in pathogenesis of diabetic nephropathy (DN). Myo-inositol oxygenase (MIOX) has been implicated in tubulointerstitial injury in the context of DN. We investigated the effect of AGEs on MIOX expression and delineated mechanisms that lead to tubulointerstitial injury. The status of MIOX, RAGE, and relevant cellular signaling pathways activated following AGE:RAGE interaction was examined in tubular cells and kidneys of AGE-BSA-treated mice. A solid-phase assay revealed an enhanced binding of RAGE with AGE-BSA, AGE-laminin, and AGE-collagen IV. The cells treated with AGE-BSA had increased MIOX activity/expression and promoter activity. This was associated with activation of various signaling kinases of phosphatidylinositol 3-kinase (PI3K)-AKT pathway and increased expression of NF-κB, transforming growth factor (TGF)-β, and fibronectin, which was negated with the treatment of MIOX/RAGE- small interfering (si) RNA. Concomitant with MIOX upregulation, there was an increased generation of reactive oxygen species (ROS), which could be abrogated with MIOX/RAGE- siRNA treatment. The kidneys of mice treated with AGE-BSA had significantly high urinary A/C ratio, upregulation of MIOX, RAGE and NF-κB, along with influx of monocytes into the tubulointerstitium, increased the expression of MCP-1, IL-6, and fibronectin and increased the generation of ROS. Such perturbations were abrogated with the concomitant treatment of inhibitors MIOX or RAGE (d-glucarate and FPS-ZM1). These studies support a role of AGE:RAGE interaction in the activation of PI3K-AKT pathway and upregulation of MIOX, with excessive generation of ROS, increased expression of NF-κB, inflammatory cytokines, TGF-β, and fibronectin. Collectively, these observations highlight the relevance of the biology of MIOX in the contribution toward tubulointerstitial injury in DN.
Collapse
Affiliation(s)
- Isha Sharma
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Rashmi S Tupe
- Biochemical Sciences Division, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth University , Pune , India
| | - Aryana K Wallner
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Yashpal S Kanwar
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
49
|
Yang D, Liu W, Ma L, Wang Y, Ma J, Jiang M, Deng X, Huang F, Yang T, Chen M. Profilin‑1 contributes to cardiac injury induced by advanced glycation end‑products in rats. Mol Med Rep 2017; 16:6634-6641. [PMID: 28901418 PMCID: PMC5865800 DOI: 10.3892/mmr.2017.7446] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 07/20/2017] [Indexed: 12/21/2022] Open
Abstract
Cardiac injury, including hypertrophy and fibrosis, induced by advanced glycation end products (AGEs) has an important function in the onset and development of diabetic cardiomyopathy. Profilin-1, a ubiquitously expressed and multifunctional actin-binding protein, has been reported to be an important mediator in cardiac hypertrophy and fibrosis. However, whether profilin-1 is involved in AGE-induced cardiac hypertrophy and fibrosis remains to be determined. Therefore, the present study aimed to investigate the function of profilin-1 in cardiac injury induced by AGEs. The model of cardiac injury was established by chronic tail vein injection of AGEs (50 mg/kg/day for 8 weeks) in Sprague-Dawley rats. Rats were randomly assigned to control, AGEs, AGEs + profilin-1 shRNA adenovirus vectors (AGEs + S)or AGEs + control adenovirus vectors (AGEs + V) groups. Profilin-1 shRNA adenovirus vectors were injected via the tail vein to knockdown profilin-1 expression at a dose of 3×109 plaque forming units every 4 weeks. Echocardiography was performed to measure cardiac contractile function. Cardiac tissues were stained with Masson's trichrome stain to evaluate ventricular remodeling. The serum levels of procollagen type III N-terminal peptide were detected by ELISA. The expression of profilin-1, receptor for AGEs (RAGE), Rho, p65, atrial natriuretic peptide, β-myosin heavy chain, matrix metalloproteinase (MMP)-2 and MMP-9 were determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and/or western blot analysis and immunohistochemistry staining. The results demonstrated that chronic injection of exogenous AGEs led to cardiac dysfunction, hypertrophy and fibrosis, as determined by echocardiography, Masson trichrome staining and the expression of associated genes. The expression of profilin-1 was markedly increased in heart tissue at the mRNA and protein level following AGE administration, as determined by RT-qPCR and western blotting, which was further confirmed by immunohistochemistry staining. Furthermore, the expression of RAGE, Rho and p65 was also increased at the protein level. Notably, knockdown of profilin-1 expression ameliorated AGE-induced cardiac injury and reduced the expression of RAGE, Rho and p65. These results indicate an important role for profilin-1 in AGE-induced cardiac injury, which may provide a novel therapeutic target for patients with diabetic heart failure.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weiwei Liu
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Liping Ma
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ya Wang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jing Ma
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Minna Jiang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xu Deng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410006, P.R. China
| | - Fang Huang
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Tianlun Yang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Meifang Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
50
|
Similarities and interactions between the ageing process and high chronic intake of added sugars. Nutr Res Rev 2017; 30:191-207. [PMID: 28511733 DOI: 10.1017/s0954422417000051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AbstractIn our societies, the proportions of elderly people and of obese individuals are increasing. Both factors are associated with high health-related costs. During obesity, many authors suggest that it is a high chronic intake of added sugars (HCIAS) that triggers the shift towards pathology. However, the majority of studies were performed in young subjects and only a few were interested in the interaction with the ageing process. Our purpose was to discuss the metabolic effects of HCIAS, compare with the effects of ageing, and evaluate how deleterious the combined action of HCIAS and ageing could be. This effect of HCIAS seems mediated by fructose, targeting the liver first, which may lead to all subsequent metabolic alterations. The first basic alterations induced by fructose are increased oxidative stress, protein glycation, inflammation, dyslipidaemia and insulin resistance. These alterations are also present during the ageing process, and are closely related to each other, one leading to the other. These basic alterations are also involved in more complex syndromes, which are also favoured by HCIAS, and present during ageing. These include non-alcoholic fatty liver disease, hypertension, neurodegenerative diseases, sarcopenia and osteoporosis. Cumulative effects of ageing and HCIAS have been seldom tested and may not always be strictly additive. Data also suggest that some of the metabolic alterations that are more prevalent during ageing could be related more with nutritional habits than to intrinsic ageing. In conclusion, it is clear that HCIAS interacts with the ageing process, accelerates the accumulation of metabolic alterations, and that it should be avoided.
Collapse
|