1
|
Li M, Chen L, Liu X, Wu Y, Chen X, Chen H, Zhong Y, Xu Y. The investigation of potential mechanism of Fuzhengkangfu Decoction against Diabetic myocardial injury based on a combined strategy of network pharmacology, transcriptomics, and experimental verification. Biomed Pharmacother 2024; 177:117048. [PMID: 38959606 DOI: 10.1016/j.biopha.2024.117048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Diabetic cardiomyopathy (DCM) is a cardiac condition resulting from myocardial damage caused by diabetes mellitus (DM), currently lacking specific therapeutic interventions. Fuzhengkangfu decoction (FZK) plays an important role in the prevention and treatment of various cardiovascular diseases. However, the efficacy and potential mechanisms of FZK are not fully understood. This study aims to investigate the protective effect and mechanisms of FZK against DCM. METHODOLOGIES Rats were given a high-calorie diet along with a low dosage of streptozotocin (STZ) to establish a rat model of DCM. The diabetic rats received FZK or normal saline subcutaneously for 12 weeks. Echocardiography was conducted to evaluate their heart function characteristics. Rat heart morphologies were assessed using Sirius Red staining and H&E staining. Transcriptome sequencing analysis and network pharmacology were used to reveal possible targets and mechanisms. Molecular docking was conducted to validate the association between the primary components of FZK and the essential target molecules. Finally, both in vitro and in vivo studies were conducted on the cardioprotective properties and mechanism of FZK. RESULTS According to the results of network pharmacology, FZK may prevent DCM by reducing oxidative stress and preventing apoptosis. Transcriptomics confirmed that FZK protected against DCM-induced myocardial fibrosis and remodelling, as predicted by network pharmacology, and suggested that FZK regulated the expression of oxidative stress and apoptosis-related proteins. Integrating network pharmacology and transcriptome analysis results revealed that the AGE-RAGE signalling pathway-associated MMP2, SLC2A1, NOX4, CCND1, and CYP1A1 might be key targets. Molecular docking showed that Poricoic acid A and 5-O-Methylvisammioside had the highest docking activities with these targets. We further conducted in vivo experiments, and the results showed that FZK significantly attenuated left ventricular remodelling, reduced myocardial fibrosis, and improved cardiac contractile function. And, our study demonstrated that FZK effectively reduced oxidative stress and apoptosis of cardiomyocytes. The data showed that Erk, NF-κB, and Caspase 3 phosphorylation was significantly inhibited, and Bcl-2/Bax was significantly increased after FZK treatment. In vitro, FZK significantly reduced AGEs-induced ROS increase and apoptosis in cardiomyocytes. Furthermore, FZK significantly inhibited the phosphorylation of Erk and NF-κB proteins and decreased the expression of MMP2. All the results confirmed that FZK inhibited the activation of the Erk/NF-κB pathway in AGE-RAGE signalling and alleviated oxidative stress and apoptosis of cardiomyocytes. In summary, we verified that FZK protects against DCM by inhibiting myocardial apoptotic remodelling through the suppression of the AGE-RAGE signalling pathway. CONCLUSION In conclusion, our research indicates that FZK demonstrates anti-cardiac dysfunction properties by reducing oxidative stress and cardiomyocyte apoptosis through the AGE-RAGE pathway in DCM, showing potential for therapeutic use.
Collapse
Affiliation(s)
- Miaofu Li
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Liuying Chen
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaohua Liu
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yirong Wu
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xuechun Chen
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Huimin Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yigang Zhong
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yizhou Xu
- Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
2
|
Graczyk P, Dach A, Dyrka K, Pawlik A. Pathophysiology and Advances in the Therapy of Cardiomyopathy in Patients with Diabetes Mellitus. Int J Mol Sci 2024; 25:5027. [PMID: 38732253 PMCID: PMC11084712 DOI: 10.3390/ijms25095027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/19/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Diabetes mellitus (DM) is known as the first non-communicable global epidemic. It is estimated that 537 million people have DM, but the condition has been properly diagnosed in less than half of these patients. Despite numerous preventive measures, the number of DM cases is steadily increasing. The state of chronic hyperglycaemia in the body leads to numerous complications, including diabetic cardiomyopathy (DCM). A number of pathophysiological mechanisms are behind the development and progression of cardiomyopathy, including increased oxidative stress, chronic inflammation, increased synthesis of advanced glycation products and overexpression of the biosynthetic pathway of certain compounds, such as hexosamine. There is extensive research on the treatment of DCM, and there are a number of therapies that can stop the development of this complication. Among the compounds used to treat DCM are antiglycaemic drugs, hypoglycaemic drugs and drugs used to treat myocardial failure. An important element in combating DCM that should be kept in mind is a healthy lifestyle-a well-balanced diet and physical activity. There is also a group of compounds-including coenzyme Q10, antioxidants and modulators of signalling pathways and inflammatory processes, among others-that are being researched continuously, and their introduction into routine therapies is likely to result in greater control and more effective treatment of DM in the future. This paper summarises the latest recommendations for lifestyle and pharmacological treatment of cardiomyopathy in patients with DM.
Collapse
Affiliation(s)
- Patryk Graczyk
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.G.); (A.D.)
| | - Aleksandra Dach
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.G.); (A.D.)
| | - Kamil Dyrka
- Department of Pediatric Endocrinology and Rheumatology, Institute of Pediatrics, Poznan University of Medical Sciences, 60-572 Poznan, Poland;
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.G.); (A.D.)
| |
Collapse
|
3
|
Hao M, Huang X, Liu X, Fang X, Li H, Lv L, Zhou L, Guo T, Yan D. Novel model predicts diastolic cardiac dysfunction in type 2 diabetes. Ann Med 2023; 55:766-777. [PMID: 36908240 PMCID: PMC10798288 DOI: 10.1080/07853890.2023.2180154] [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: 10/07/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
OBJECTIVE Diabetes mellitus complicated with heart failure has high mortality and morbidity, but no reliable diagnoses and treatments are available. This study aimed to develop and verify a new model nomogram based on clinical parameters to predict diastolic cardiac dysfunction in patients with Type 2 diabetes mellitus (T2DM). METHODS 3030 patients with T2DM underwent Doppler echocardiography at the First Affiliated Hospital of Shenzhen University between January 2014 and December 2021. The patients were divided into the training dataset (n = 1701) and the verification dataset (n = 1329). In this study, a predictive diastolic cardiac dysfunction nomogram is developed using multivariable logical regression analysis, which contains the candidates selected in a minor absolute shrinkage and selection operator regression model. Discrimination in the prediction model was assessed using the area under the receiver operating characteristic curve (AUC-ROC). The calibration curve was applied to evaluate the calibration of the alignment nomogram, and the clinical decision curve was used to determine the clinical practicability of the alignment map. The verification dataset was used to evaluate the prediction model's performance. RESULTS A multivariable model that included age, body mass index (BMI), triglyceride (TG), creatine phosphokinase isoenzyme (CK-MB), serum sodium (Na), and urinary albumin/creatinine ratio (UACR) was presented as the nomogram. We obtained the model for estimating diastolic cardiac dysfunction in patients with T2DM. The AUC-ROC of the training dataset in our model was 0.8307, with 95% CI of 0.8109-0.8505. Similar to the results obtained with the training dataset, the AUC-ROC of the verification dataset in our model was 0.8083, with 95% CI of 0.7843-0.8324, thus demonstrating robust. The function of the predictive model was as follows: Diastolic Dysfunction = -4.41303 + 0.14100*Age(year)+0.10491*BMI (kg/m2) +0.12902*TG (mmol/L) +0.03970*CK-MB (ng/mL) -0.03988*Na(mmol/L) +0.65395 * (UACR > 30 mg/g) + 1.10837 * (UACR > 300 mg/g). The calibration plot diagram of predicted probabilities against observed DCM rates indicated excellent concordance. Decision curve analysis demonstrated that the novel nomogram was clinically useful. CONCLUSION Diastolic cardiac dysfunction in patients with T2DM can be predicted by clinical parameters. Our prediction model may represent an effective tool for large-scale epidemiological study of diastolic cardiac dysfunction in T2DM patients and provide a reliable method for early screening of T2DM patients with cardiac complications.KEY MESSAGESThis study used clinical parameters to predict diastolic cardiac dysfunction in patients with T2DM. This study established a nomogram for predicting diastolic cardiac dysfunction by multivariate logical regression analysis. Our predictive model can be used as an effective tool for large-scale epidemiological study of diastolic cardiac dysfunction in patients with T2DM and provides a reliable method for early screening of cardiac complications in patients with T2DM.
Collapse
Affiliation(s)
- Mingyu Hao
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaohong Huang
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
- Guangzhou Medical University, Guangzhou, China
| | - Xueting Liu
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Xiaokang Fang
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Haiyan Li
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Lingbo Lv
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Liming Zhou
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Tiecheng Guo
- Chiwan Community Health Service Centre, Shenzhen, China
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Allemann MS, Lee P, Beer JH, Saeedi Saravi SS. Targeting the redox system for cardiovascular regeneration in aging. Aging Cell 2023; 22:e14020. [PMID: 37957823 PMCID: PMC10726899 DOI: 10.1111/acel.14020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular aging presents a formidable challenge, as the aging process can lead to reduced cardiac function and heightened susceptibility to cardiovascular diseases. Consequently, there is an escalating, unmet medical need for innovative and effective cardiovascular regeneration strategies aimed at restoring and rejuvenating aging cardiovascular tissues. Altered redox homeostasis and the accumulation of oxidative damage play a pivotal role in detrimental changes to stem cell function and cellular senescence, hampering regenerative capacity in aged cardiovascular system. A mounting body of evidence underscores the significance of targeting redox machinery to restore stem cell self-renewal and enhance their differentiation potential into youthful cardiovascular lineages. Hence, the redox machinery holds promise as a target for optimizing cardiovascular regenerative therapies. In this context, we delve into the current understanding of redox homeostasis in regulating stem cell function and reprogramming processes that impact the regenerative potential of the cardiovascular system. Furthermore, we offer insights into the recent translational and clinical implications of redox-targeting compounds aimed at enhancing current regenerative therapies for aging cardiovascular tissues.
Collapse
Affiliation(s)
- Meret Sarah Allemann
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Pratintip Lee
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Jürg H. Beer
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of CardiologyUniversity Hospital Zurich, University of ZurichSchlierenSwitzerland
| |
Collapse
|
5
|
Bao J, Gao Z, Hu Y, Ye L, Wang L. Transient receptor potential vanilloid type 1: cardioprotective effects in diabetic models. Channels (Austin) 2023; 17:2281743. [PMID: 37983306 PMCID: PMC10761101 DOI: 10.1080/19336950.2023.2281743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
Cardiovascular disease, especially heart failure (HF) is the leading cause of death in patients with diabetes. Individuals with diabetes are prone to a special type of cardiomyopathy called diabetic cardiomyopathy (DCM), which cannot be explained by heart diseases such as hypertension or coronary artery disease, and can contribute to HF. Unfortunately, the current treatment strategy for diabetes-related cardiovascular complications is mainly to control blood glucose levels; nonetheless, the improvement of cardiac structure and function is not ideal. The transient receptor potential cation channel subfamily V member 1 (TRPV1), a nonselective cation channel, has been shown to be universally expressed in the cardiovascular system. Increasing evidence has shown that the activation of TRPV1 channel has a potential protective influence on the cardiovascular system. Numerous studies show that activating TRPV1 channels can improve the occurrence and progression of diabetes-related complications, including cardiomyopathy; however, the specific mechanisms and effects are unclear. In this review, we summarize that TRPV1 channel activation plays a protective role in the heart of diabetic models from oxidation/nitrification stress, mitochondrial function, endothelial function, inflammation, and cardiac energy metabolism to inhibit the occurrence and progression of DCM. Therefore, TRPV1 may become a latent target for the prevention and treatment of diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhicheng Gao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Lee TI, Trang NN, Lee TW, Higa S, Kao YH, Chen YC, Chen YJ. Ketogenic Diet Regulates Cardiac Remodeling and Calcium Homeostasis in Diabetic Rat Cardiomyopathy. Int J Mol Sci 2023; 24:16142. [PMID: 38003332 PMCID: PMC10671812 DOI: 10.3390/ijms242216142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
A ketogenic diet (KD) might alleviate patients with diabetic cardiomyopathy. However, the underlying mechanism remains unclear. Myocardial function and arrhythmogenesis are closely linked to calcium (Ca2+) homeostasis. We investigated the effects of a KD on Ca2+ homeostasis and electrophysiology in diabetic cardiomyopathy. Male Wistar rats were created to have diabetes mellitus (DM) using streptozotocin (65 mg/kg, intraperitoneally), and subsequently treated for 6 weeks with either a normal diet (ND) or a KD. Our electrophysiological and Western blot analyses assessed myocardial Ca2+ homeostasis in ventricular preparations in vivo. Unlike those on the KD, DM rats treated with an ND exhibited a prolonged QTc interval and action potential duration. Compared to the control and DM rats on the KD, DM rats treated with an ND also showed lower intracellular Ca2+ transients, sarcoplasmic reticular Ca2+ content, sodium (Na+)-Ca2+ exchanger currents (reverse mode), L-type Ca2+ contents, sarcoplasmic reticulum ATPase contents, Cav1.2 contents. Furthermore, these rats exhibited elevated ratios of phosphorylated to total proteins across multiple Ca2+ handling proteins, including ryanodine receptor 2 (RyR2) at serine 2808, phospholamban (PLB)-Ser16, and calmodulin-dependent protein kinase II (CaMKII). Additionally, DM rats treated with an ND demonstrated a higher frequency and incidence of Ca2+ leak, cytosolic reactive oxygen species, Na+/hydrogen-exchanger currents, and late Na+ currents than the control and DM rats on the KD. KD treatment may attenuate the effects of DM-dysregulated Na+ and Ca2+ homeostasis, contributing to its cardioprotection in DM.
Collapse
Affiliation(s)
- Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (T.-I.L.); (T.-W.L.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | | | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (T.-I.L.); (T.-W.L.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Makiminato Urasoe City, Okinawa 901-2131, Japan;
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| |
Collapse
|
7
|
Roohi TF, Faizan S, Parray ZA, Baig MDAI, Mehdi S, Kinattingal N, Krishna KL. Beyond Glucose: The Dual Assault of Oxidative and ER Stress in Diabetic Disorders. High Blood Press Cardiovasc Prev 2023; 30:513-531. [PMID: 38041772 DOI: 10.1007/s40292-023-00611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Diabetes mellitus, a prevalent global health concern, is characterized by hyperglycemia. However, recent research reveals a more intricate landscape where oxidative stress and endoplasmic reticulum (ER) stress orchestrate a dual assault, profoundly impacting diabetic disorders. This review elucidates the interplay between these two stress pathways and their collective consequences on diabetes. Oxidative stress emanates from mitochondria, where reactive oxygen species (ROS) production spirals out of control, leading to cellular damage. We explore ROS-mediated signaling pathways, which trigger β-cell dysfunction, insulin resistance, and endothelial dysfunction the quintessential features of diabetes. Simultaneously, ER stress unravels, unveiling how protein folding disturbances activate the unfolded protein response (UPR). We dissect the UPR's dual role, oscillating between cellular adaptation and apoptosis, significantly influencing pancreatic β-cells and peripheral insulin-sensitive tissues. Crucially, this review exposes the synergy between oxidative and ER stress pathways. ROS-induced UPR activation and ER stress-induced oxidative stress create a detrimental feedback loop, exacerbating diabetic complications. Moreover, we spotlight promising therapeutic strategies that target both stress pathways. Antioxidants, molecular chaperones, and novel pharmacological agents offer potential avenues for diabetes management. As the global diabetes burden escalates, comprehending the dual assault of oxidative and ER stress is paramount. This review not only unveils the intricate molecular mechanisms governing diabetic pathophysiology but also advocates a holistic therapeutic approach. By addressing both stress pathways concurrently, we may forge innovative solutions for diabetic disorders, ultimately alleviating the burden of this pervasive health issue.
Collapse
Affiliation(s)
- Tamsheel Fatima Roohi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Syed Faizan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Zahoor Ahmad Parray
- Department of Chemistry, Indian Institute of Technology (IIT) Delhi, Hauz Khas Campus, New Delhi, 110016, India
| | - M D Awaise Iqbal Baig
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Seema Mehdi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Nabeel Kinattingal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - K L Krishna
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India.
| |
Collapse
|
8
|
Al-Masri A. Apoptosis and long non-coding RNAs: Focus on their roles in Heart diseases. Pathol Res Pract 2023; 251:154889. [PMID: 38238070 DOI: 10.1016/j.prp.2023.154889] [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: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 01/23/2024]
Abstract
Heart disease is one of the principal death reasons around the world and there is a growing requirement to discover novel healing targets that have the potential to avert or manage these illnesses. On the other hand, apoptosis is a strongly controlled, cell removal procedure that has a crucial part in numerous cardiac problems, such as reperfusion injury, MI (myocardial infarction), consecutive heart failure, and inflammation of myocardium. Completely comprehending the managing procedures of cell death signaling is critical as it is the primary factor that influences patient mortality and morbidity, owing to cardiomyocyte damage. Indeed, the prevention of heart cell death appears to be a viable treatment approach for heart illnesses. According to current researches, a number of long non-coding RNAs cause the heart cells death via different methods that are embroiled in controlling the activity of transcription elements, the pathways that signals transmission within cells, small miRNAs, and the constancy of proteins. When there is too much cell death in the heart, it can cause problems like reduced blood flow, heart damage after restoring blood flow, heart disease in diabetics, and changes in the heart after reduced blood flow. Therefore, studying how lncRNAs control apoptosis could help us find new treatments for heart diseases. In this review, we present recent discoveries about how lncRNAs are involved in causing cell death in different cardiovascular diseases.
Collapse
Affiliation(s)
- Abeer Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
9
|
Xiao SL, Bober E, Kassianides X, Medici F, Xiao HB. Diabetic cardiomyopathy: an educational review. THE BRITISH JOURNAL OF CARDIOLOGY 2023; 30:18. [PMID: 38911684 PMCID: PMC11190836 DOI: 10.5837/bjc.2023.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
This educational review provides information about the epidemiology of diabetes and heart failure (diabetic cardiomyopathy) and the challenges in diagnosis and screening. Details on how to investigate patients with imaging and other modalities are discussed, as well as an update regarding the efficacy and safety of novel agents for treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Su-Lee Xiao
- Specialist Registrar (ST5) Geriatric Medicine Charing Cross Hospital, Fulham Palace Road, London W6 8RF
| | - Emilia Bober
- Foundation Year 1 Doctor Northwick Park Hospital, Watford Road, Harrow, Middlesex, HA1 3UJ
| | - Xenophon Kassianides
- Specialty Registar in General Practice Hull University Teaching Hospitals NHS Trust and the Hull York Medical School, Anlaby Road, Hull, HU3 2JZ
| | | | - Han B Xiao
- Consultant Cardiologist Homerton Hospital, Homerton Row, London, E9 6SR
| |
Collapse
|
10
|
Emerging Therapy for Diabetic Cardiomyopathy: From Molecular Mechanism to Clinical Practice. Biomedicines 2023; 11:biomedicines11030662. [PMID: 36979641 PMCID: PMC10045486 DOI: 10.3390/biomedicines11030662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/24/2023] Open
Abstract
Diabetic cardiomyopathy is characterized by abnormal myocardial structure or performance in the absence of coronary artery disease or significant valvular heart disease in patients with diabetes mellitus. The spectrum of diabetic cardiomyopathy ranges from subtle myocardial changes to myocardial fibrosis and diastolic function and finally to symptomatic heart failure. Except for sodium–glucose transport protein 2 inhibitors and possibly bariatric and metabolic surgery, there is currently no specific treatment for this distinct disease entity in patients with diabetes. The molecular mechanism of diabetic cardiomyopathy includes impaired nutrient-sensing signaling, dysregulated autophagy, impaired mitochondrial energetics, altered fuel utilization, oxidative stress and lipid peroxidation, advanced glycation end-products, inflammation, impaired calcium homeostasis, abnormal endothelial function and nitric oxide production, aberrant epidermal growth factor receptor signaling, the activation of the renin–angiotensin–aldosterone system and sympathetic hyperactivity, and extracellular matrix accumulation and fibrosis. Here, we summarize several important emerging treatments for diabetic cardiomyopathy targeting specific molecular mechanisms, with evidence from preclinical studies and clinical trials.
Collapse
|
11
|
Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications. Antioxidants (Basel) 2023; 12:antiox12010123. [PMID: 36670985 PMCID: PMC9855127 DOI: 10.3390/antiox12010123] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/10/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The global prevalence of diabetes continues to increase partly due to rapid urbanization and an increase in the aging population. Consequently, this is associated with a parallel increase in the prevalence of diabetic vascular complications which significantly worsen the burden of diabetes. For these diabetic vascular complications, there is still an unmet need for safe and effective alternative/adjuvant therapeutic interventions. There is also an increasing urge for therapeutic options to come from natural products such as plants. Hyperglycemia-induced oxidative stress is central to the development of diabetes and diabetic complications. Furthermore, oxidative stress-induced inflammation and insulin resistance are central to endothelial damage and the progression of diabetic complications. Human and animal studies have shown that polyphenols could reduce oxidative stress, hyperglycemia, and prevent diabetic complications including diabetic retinopathy, diabetic nephropathy, and diabetic peripheral neuropathy. Part of the therapeutic effects of polyphenols is attributed to their modulatory effect on endogenous antioxidant systems. This review attempts to summarize the established effects of polyphenols on endogenous antioxidant systems from the literature. Moreover, potential therapeutic strategies for harnessing the potential benefits of polyphenols for diabetic vascular complications are also discussed.
Collapse
|
12
|
Syed AA, Reza MI, Shafiq M, Kumariya S, Katekar R, Hanif K, Gayen JR. Cissus quadrangularis extract mitigates diabetic cardiomyopathy by inhibiting RAAS activation, inflammation, and oxidative stress. Biomarkers 2022; 27:743-752. [PMID: 35896310 DOI: 10.1080/1354750x.2022.2107703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
IntroductionDiabetic cardiomyopathy (DCM) is an age-related disease, and its progression is accompanied by hyperglycemia, cardiac dysfunction, and myocardial structural and functional abnormalities. Cissus quadrangularis, a traditional medicinal plant, contains polyphenols, flavonoids, phytosterols, carbohydrates, and ascorbic acid. It is used to treat osteoporosis, asthma, haemorrhoids, and menstrual disorders. In the current research, we have investigated the effect of ethanolic extract of C. quadrangularis (EECQ) against a high-fat diet (HFD)/streptozotocin-induced DCM by estimating cardiac biomarkers, inflammatory markers and ROS production.Material and methodsRats were fed with an HFD for 12 weeks, followed by single-shot low-dose streptozotocin (35mg/kg; i.p.). The treatment was performed by EECQ (200 mg/kg/day, orally) for six weeks. ResultsThe extract EECQ improves glucose, insulin tolerance tests, and hypercholesteremia. DCM is characterized by cardiac dysfunction, cardiac biomarkers CKMB, and LDH, which were attenuated by the EECQ treatment. The hypertrophic biomarker ANP, BNP expression and cardiomyocyte surface area were decreased by EECQ. Moreover, EECQ also alleviated the biomarkers Angiotensin II and renin level. EECQ also reduced oxidative stress, ROS production and cardiac inflammation.ConclusionThus, these findings suggested that EECQ could be used as a possible therapeutic regiment to treat DCM.
Collapse
Affiliation(s)
- Anees Ahmed Syed
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Mohammad Irshad Reza
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India
| | - Mohammed Shafiq
- Pharmacology Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Sanjana Kumariya
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India
| | - Roshan Katekar
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Kashif Hanif
- Pharmacology Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India.,Pharmacology Division, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| |
Collapse
|
13
|
Gao G, Fu L, Xu Y, Tao L, Guo T, Fang G, Zhang G, Wang S, Qin T, Luo P, Shen X. Cyclovirobuxine D Ameliorates Experimental Diabetic Cardiomyopathy by Inhibiting Cardiomyocyte Pyroptosis via NLRP3 in vivo and in vitro. Front Pharmacol 2022; 13:906548. [PMID: 35865939 PMCID: PMC9294384 DOI: 10.3389/fphar.2022.906548] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the common complications of diabetic patients, which can induce myocardial hypertrophy, cardiac fibrosis, and heart failure. Growing evidence has shown that the occurrence and development of DCM are accompanied by pyroptosis which is an NLRP3-mediated intense inflammatory cell death. Cyclovirobuxine D (CVB-D) has been shown to significantly ameliorate DCM and anti-inflammatory effects associated with cardiomyopathy, but it is unclear whether it has an effect on cardiomyocyte pyroptosis accompanying DCM. Therefore, the purpose of the present study was to explore the ameliorating effect of CVB-D on cardiomyocyte pyroptosis associated with DCM and its molecular regulation mechanism. Type 2 diabetes in C57BL/6 mice was reproduced by the high-fat and high-glucose diet (HFD) combined with low-dose streptozotocin (STZ). The characteristics of DCM were evaluated by cardiac ultrasonography, serum detection, and histopathological staining. The results suggested that CVB-D could significantly alleviate the cardiac pathology of DCM. Then, we explored the mechanism of CVB-D on primary neonatal rat cardiomyocyte (PNRCM) injury with high glucose (HG) in vitro to simulate the physiological environment of DCM. Preincubation with CVB-D could significantly increase cell viability, attenuate cytopathological changes and inhibit the expression levels of pyroptosis-related proteins. Further research found that the myocardial improvement effect of CVB-D was related to its inhibition of NLRP3 expression. In conclusion, our data suggest that CVB-D can ameliorate DCM by inhibiting cardiomyocyte pyroptosis via NLRP3, providing a novel molecular target for CVB-D clinical application.
Collapse
Affiliation(s)
- Ge Gao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Lingyun Fu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Yini Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Ting Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Guanqin Fang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Guangqiong Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Shengquan Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Ti Qin
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Peng Luo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- *Correspondence: Peng Luo, ; Xiangchun Shen,
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang, China
- *Correspondence: Peng Luo, ; Xiangchun Shen,
| |
Collapse
|
14
|
Krishnan A, Sharma H, Yuan D, Trollope AF, Chilton L. The Role of Epicardial Adipose Tissue in the Development of Atrial Fibrillation, Coronary Artery Disease and Chronic Heart Failure in the Context of Obesity and Type 2 Diabetes Mellitus: A Narrative Review. J Cardiovasc Dev Dis 2022; 9:jcdd9070217. [PMID: 35877579 PMCID: PMC9318726 DOI: 10.3390/jcdd9070217] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 12/07/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a significant burden globally and are especially prevalent in obese and/or diabetic populations. Epicardial adipose tissue (EAT) surrounding the heart has been implicated in the development of CVDs as EAT can shift from a protective to a maladaptive phenotype in diseased states. In diabetic and obese patients, an elevated EAT mass both secretes pro-fibrotic/pro-inflammatory adipokines and forms intramyocardial fibrofatty infiltrates. This narrative review considers the proposed pathophysiological roles of EAT in CVDs. Diabetes is associated with a disordered energy utilization in the heart, which promotes intramyocardial fat and structural remodeling. Fibrofatty infiltrates are associated with abnormal cardiomyocyte calcium handling and repolarization, increasing the probability of afterdepolarizations. The inflammatory phenotype also promotes lateralization of connexin (Cx) proteins, undermining unidirectional conduction. These changes are associated with conduction heterogeneity, together creating a substrate for atrial fibrillation (AF). EAT is also strongly implicated in coronary artery disease (CAD); inflammatory adipokines from peri-vascular fat can modulate intra-luminal homeostasis through an “outside-to-inside” mechanism. EAT is also a significant source of sympathetic neurotransmitters, which promote progressive diastolic dysfunction with eventual cardiac failure. Further investigations on the behavior of EAT in diabetic/obese patients with CVD could help elucidate the pathogenesis and uncover potential therapeutic targets.
Collapse
Affiliation(s)
- Anirudh Krishnan
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Harman Sharma
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Daniel Yuan
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Alexandra F. Trollope
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia;
| | - Lisa Chilton
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia
- Correspondence:
| |
Collapse
|
15
|
Pofi R, Giannetta E, Feola T, Galea N, Barbagallo F, Campolo F, Badagliacca R, Barbano B, Ciolina F, Defeudis G, Filardi T, Sesti F, Minnetti M, Vizza CD, Pasqualetti P, Caboni P, Carbone I, Francone M, Catalano C, Pozzilli P, Lenzi A, Venneri MA, Gianfrilli D, Isidori AM. Sex-specific effects of daily tadalafil on diabetic heart kinetics in RECOGITO, a randomized, double-blind, placebo-controlled trial. Sci Transl Med 2022; 14:eabl8503. [PMID: 35704597 DOI: 10.1126/scitranslmed.abl8503] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclic GMP-phosphodiesterase type 5 (PDE5) inhibition has been shown to counteract maladaptive cardiac changes triggered by diabetes in some but not all studies. We performed a single-center, 20-week, double-blind, randomized, placebo-controlled trial (NCT01803828) to assess sex differences in cardiac remodeling after PDE5 inhibition in patients with diabetic cardiomyopathy. A total of 122 men and women (45 to 80 years) with long-duration (>3 years) and well-controlled type 2 diabetes mellitus (T2DM; HbA1c < 86 mmol/mol) were selected according to echocardiographic signs of cardiac remodeling. Patients were randomly assigned (1:1) to placebo or oral tadalafil (20 mg, once daily). The primary outcome was to evaluate sex differences in cardiac torsion change. Secondary outcomes were changes in cardiovascular, metabolic, immune, and renal function. At 20 weeks, the treatment-by-sex interaction documented an improvement in cardiac torsion (-3.40°, -5.96; -0.84, P = 0.011) and fiber shortening (-1.19%, -2.24; -0.14, P = 0.027) in men but not women. The primary outcome could not be explained by differences in cGMP concentrations or tadalafil pharmacodynamics. In both sexes, tadalafil improved hsa-miR-199-5p expression, biomarkers of cardiovascular remodeling, albuminuria, renal artery resistive index, and circulating Klotho concentrations. Immune cell profiling revealed an improvement in low-grade chronic inflammation: Classic CD14++CD16- monocytes reduced, and Tie2+ monocytes increased. Nine patients (14.5%) had minor adverse reactions after tadalafil administration. Continuous PDE5 inhibition could offer a strategy to target cardiorenal complications of T2DM, with sex- and tissue-specific responses. Further studies are needed to confirm Klotho and hsa-miR-199-5p as markers for T2DM complications.
Collapse
Affiliation(s)
- Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.,Neuroendocrinology, Neuromed Institute, IRCCS, 86077 Pozzilli (IS), Italy
| | - Nicola Galea
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Roberto Badagliacca
- Department of Cardiovascular and Respiratory Diseases, Sapienza University of Rome, 00161 Rome, Italy
| | - Biagio Barbano
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Ciolina
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giuseppe Defeudis
- Unit of Endocrinology and Diabetes, Department of Medicine, University Campus Bio-Medico di Roma, 00161 Rome, Italy
| | - Tiziana Filardi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Franz Sesti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marianna Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Carmine D Vizza
- Department of Cardiovascular and Respiratory Diseases, Sapienza University of Rome, 00161 Rome, Italy
| | - Patrizio Pasqualetti
- Medical Statistics and Information Technology, AFaR, Fatebenefratelli Hospital, 00161 Rome, Italy
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Iacopo Carbone
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Francone
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Carlo Catalano
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Paolo Pozzilli
- Unit of Endocrinology and Diabetes, Department of Medicine, University Campus Bio-Medico di Roma, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
16
|
Lin H, Wang Z, Luo Y, Lin Z, Hong G, Deng K, Huang P, Shen Y. Non/mini-invasive monitoring of diabetes-induced myocardial damage by Fourier transform infrared spectroscopy: Evidence from biofluids. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166445. [PMID: 35577177 DOI: 10.1016/j.bbadis.2022.166445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 11/26/2022]
Abstract
Early identification of diabetic cardiomyopathy (DCM) can help clinicians develop targeted treatment plans and forensic pathologists make accurate postmortem diagnoses. In the present study, diabetes-induced metabolic abnormalities in the myocardium and biofluids (plasma, urine, and saliva) of db/db mice of various ages (7, 12, and 21 weeks) were investigated by attenuated total reflection (ATR)-Fourier transform infrared (FTIR) spectroscopy. The results indicated that the diabetic and control groups had significantly different changes in the function groups of lipids, phosphate macromolecules (mostly nucleic acids), protein compositions and conformations, and carbohydrates (primarily glucose) in the myocardium and biofluids. The prediction model for quantifying DCM severity was developed on db/db mice's myocardial spectra using a genetic algorithm (GA)-partial least squares (PLS) regression method. Following that, the linear correlations between the predicted values for DCM severity and spectra for db/db biofluids were evaluated using the GA-PLS regression algorithm. The results showed there were good linear correlations between the predicted values for DCM severity and spectra for plasma (R2 = 0.929), saliva (R2 = 0.967), urine (R2 = 0.954), and combination of plasma and saliva (R2 = 0.980). This study provides a novel perspective on detecting diabetes-related biofluid and cardiac metabolic abnormalities and demonstrates the potential of biofluid infrared spectro-diagnostic models for non/mini-invasive assessment of DCM.
Collapse
Affiliation(s)
- Hancheng Lin
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhimin Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yiwen Luo
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Science, Ministry of Justice, PRC, Shanghai 200063, China
| | - Zijie Lin
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Guanghui Hong
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Kaifei Deng
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Science, Ministry of Justice, PRC, Shanghai 200063, China
| | - Ping Huang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Science, Ministry of Justice, PRC, Shanghai 200063, China.
| | - Yiwen Shen
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
17
|
Hao M, Deng J, Huang X, Li H, Ou H, Cai X, She J, Liu X, Chen L, Chen S, Liu W, Yan D. Metabonomic Characteristics of Myocardial Diastolic Dysfunction in Type 2 Diabetic Cardiomyopathy Patients. Front Physiol 2022; 13:863347. [PMID: 35651872 PMCID: PMC9150260 DOI: 10.3389/fphys.2022.863347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the most essential cardiovascular complications in diabetic patients associated with glucose and lipid metabolism disorder, fibrosis, oxidative stress, and inflammation in cardiomyocytes. Despite increasing research on the molecular pathogenesis of DCM, it is still unclear whether metabolic pathways and alterations are probably involved in the development of DCM. This study aims to characterize the metabolites of DCM and to identify the relationship between metabolites and their biological processes or biological states through untargeted metabolic profiling. UPLC-MS/MS was applied to profile plasma metabolites from 78 patients with diabetes (39 diabetes with DCM and 39 diabetes without DCM as controls). A total of 2,806 biochemical were detected. Compared to those of DM patients, 78 differential metabolites in the positive-ion mode were identified in DCM patients, including 33 up-regulated and 45 down-regulated metabolites; however, there were only six differential metabolites identified in the negative mode including four up-regulated and two down-regulated metabolites. Alterations of several serum metabolites, including lipids and lipid-like molecules, organic acids and derivatives, organic oxygen compounds, benzenoids, phenylpropanoids and polyketides, and organoheterocyclic compounds, were associated with the development of DCM. KEGG enrichment analysis showed that there were three signaling pathways (metabolic pathways, porphyrin, chlorophyll metabolism, and lysine degradation) that were changed in both negative- and positive-ion modes. Our results demonstrated that differential metabolites and lipids have specific effects on DCM. These results expanded our understanding of the metabolic characteristics of DCM and may provide a clue in the future investigation of reducing the incidence of DCM. Furthermore, the metabolites identified here may provide clues for clinical management and the development of effective drugs.
Collapse
Affiliation(s)
- Mingyu Hao
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianxin Deng
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
- *Correspondence: Jianxin Deng, , ; Wenlan Liu, ; Dewen Yan,
| | - Xiaohong Huang
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Haiyan Li
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Huiting Ou
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Xiangsheng Cai
- Institute of Translational Medicine, University of Chinese Academy of Science-Shenzhen Hospital, Shenzhen, China
| | - Jiajie She
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xueting Liu
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Ling Chen
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Shujuan Chen
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Wenlan Liu
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People’s Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, China
- *Correspondence: Jianxin Deng, , ; Wenlan Liu, ; Dewen Yan,
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
- *Correspondence: Jianxin Deng, , ; Wenlan Liu, ; Dewen Yan,
| |
Collapse
|
18
|
Caturano A, Vetrano E, Galiero R, Salvatore T, Docimo G, Epifani R, Alfano M, Sardu C, Marfella R, Rinaldi L, Sasso FC. Cardiac Hypertrophy: From Pathophysiological Mechanisms to Heart Failure Development. Rev Cardiovasc Med 2022; 23:165. [PMID: 39077592 PMCID: PMC11273913 DOI: 10.31083/j.rcm2305165] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 07/31/2024] Open
Abstract
Cardiac hypertrophy develops in response to increased workload to reduce ventricular wall stress and maintain function and efficiency. Pathological hypertrophy can be adaptive at the beginning. However, if the stimulus persists, it may progress to ventricular chamber dilatation, contractile dysfunction, and heart failure, resulting in poorer outcome and increased social burden. The main pathophysiological mechanisms of pathological hypertrophy are cell death, fibrosis, mitochondrial dysfunction, dysregulation of Ca 2 + -handling proteins, metabolic changes, fetal gene expression reactivation, impaired protein and mitochondrial quality control, altered sarcomere structure, and inadequate angiogenesis. Diabetic cardiomyopathy is a condition in which cardiac pathological hypertrophy mainly develop due to insulin resistance and subsequent hyperglycaemia, associated with altered fatty acid metabolism, altered calcium homeostasis and inflammation. In this review, we summarize the underlying molecular mechanisms of pathological hypertrophy development and progression, which can be applied in the development of future novel therapeutic strategies in both reversal and prevention.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Giovanni Docimo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaella Epifani
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Maria Alfano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| |
Collapse
|
19
|
Plin5, a New Target in Diabetic Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2122856. [PMID: 35509833 PMCID: PMC9060988 DOI: 10.1155/2022/2122856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
Abstract
Abnormal lipid accumulation is commonly observed in diabetic cardiomyopathy (DC), which can create a lipotoxic microenvironment and damage cardiomyocytes. Lipid toxicity is an important pathogenic factor due to abnormal lipid accumulation in DC. As a lipid droplet (LD) decomposition barrier, Plin5 can protect LDs from lipase decomposition and regulate lipid metabolism, which is involved in the occurrence and development of cardiovascular diseases. In recent years, studies have shown that Plin5 expression is involved in the pathogenesis of DC lipid toxicity, such as oxidative stress, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and insulin resistance (IR) and has become a key target of DC research. Therefore, understanding the relationship between Plin5 and DC progression as well as the mechanism of this process is crucial for developing new therapeutic approaches and exploring new therapeutic targets. This review is aimed at exploring the latest findings and roles of Plin5 in lipid metabolism and DC-related pathogenesis, to explore possible clinical intervention approaches.
Collapse
|
20
|
Gatica D, Chiong M, Lavandero S, Klionsky DJ. The role of autophagy in cardiovascular pathology. Cardiovasc Res 2022; 118:934-950. [PMID: 33956077 PMCID: PMC8930074 DOI: 10.1093/cvr/cvab158] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
Macroautophagy/autophagy is a conserved catabolic recycling pathway in which cytoplasmic components are sequestered, degraded, and recycled to survive various stress conditions. Autophagy dysregulation has been observed and linked with the development and progression of several pathologies, including cardiovascular diseases, the leading cause of death in the developed world. In this review, we aim to provide a broad understanding of the different molecular factors that govern autophagy regulation and how these mechanisms are involved in the development of specific cardiovascular pathologies, including ischemic and reperfusion injury, myocardial infarction, cardiac hypertrophy, cardiac remodelling, and heart failure.
Collapse
Affiliation(s)
- Damián Gatica
- Department of Molecular, Cellular and Developmental Biology, Life Sciences Institute, University of Michigan, 210 Washtenaw Ave, Ann Arbor, MI 48109-2216, USA
| | - Mario Chiong
- Department of Biochemistry and Molecular Biology, Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Olivos 1007, Independencia, Santiago 8380492, Chile
| | - Sergio Lavandero
- Department of Biochemistry and Molecular Biology, Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Olivos 1007, Independencia, Santiago 8380492, Chile
- Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), 926 JF Gonzalez, Santiago 7860201, Chile
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-8573, USA
| | - Daniel J Klionsky
- Department of Molecular, Cellular and Developmental Biology, Life Sciences Institute, University of Michigan, 210 Washtenaw Ave, Ann Arbor, MI 48109-2216, USA
| |
Collapse
|
21
|
Potere N, Del Buono MG, Vecchié A, Porreca E, Abbate A, Dentali F, Bonaventura A. Diabetes mellitus and heart failure: an update on pathophysiology and therapy. Minerva Cardiol Angiol 2022; 70:344-356. [PMID: 35212512 DOI: 10.23736/s2724-5683.22.05967-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Diabetes mellitus (DM) is frequent among heart failure (HF) patients with a further projected increase in prevalence in next years. DM promotes the development of both HF with reduced (HFrEF) and preserved ejection fraction (HFpEF) through different mechanisms. As the general prevalence of both DM and HF is growing worldwide, it is important to define the pathophysiologic mechanisms driving the development of HF in DM patients. These include changes in the cardiac metabolism, mitochondrial dysfunction, impairment in insulin signaling, maladaptive inflammation, coronary microvascular dysfunction, endoplasmic reticulum stress, autophagy suppression, and structural changes, among the main ones. In recent years, novel glucose-lowering treatments, especially sodium-glucose cotransporter 2 inhibitors (SGLT-2is), have shown a strikingly positive impact on the natural history of HF. This has led to a progressive change in choosing SGLT-2is in DM patients at high risk for CV disease, supported by recent guidelines. The knowledge about novel pathophysiological mechanisms linking DM and HF may open the way to the development of new targeted therapies in the future. In this review, we will summarize general aspects dealing with incidence, prevalence, and pathophysiology of DM in HF patients. As well, we discuss the therapeutic targets to reduce the disease burden and the current evidence of glucose-lowering drugs in patients with DM and HF.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences and Department of Innovative Technologies in Medicine and Dentistry, G. D'Annunzio University, Chieti, Italy
| | - Marco G Del Buono
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA.,Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Alessandra Vecchié
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Ettore Porreca
- Department of Medicine and Ageing Sciences and Department of Innovative Technologies in Medicine and Dentistry, G. D'Annunzio University, Chieti, Italy
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy -
| |
Collapse
|
22
|
Pavez-Giani MG, Cyganek L. Recent Advances in Modeling Mitochondrial Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Front Cell Dev Biol 2022; 9:800529. [PMID: 35083221 PMCID: PMC8784695 DOI: 10.3389/fcell.2021.800529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022] Open
Abstract
Around one third of patients with mitochondrial disorders develop a kind of cardiomyopathy. In these cases, severity is quite variable ranging from asymptomatic status to severe manifestations including heart failure, arrhythmias, and sudden cardiac death. ATP is primarily generated in the mitochondrial respiratory chain via oxidative phosphorylation by utilizing fatty acids and carbohydrates. Genes in both the nuclear and the mitochondrial DNA encode components of this metabolic route and, although mutations in these genes are extremely rare, the risk to develop cardiac symptoms is significantly higher in this patient cohort. Additionally, infants with cardiovascular compromise in mitochondrial deficiency display a worse late survival compared to patients without cardiac symptoms. At this point, the mechanisms behind cardiac disease progression related to mitochondrial gene mutations are poorly understood and current therapies are unable to substantially restore the cardiac performance and to reduce the disease burden. Therefore, new strategies are needed to uncover the pathophysiological mechanisms and to identify new therapeutic options for mitochondrial cardiomyopathies. Here, human induced pluripotent stem cell (iPSC) technology has emerged to provide a suitable patient-specific model system by recapitulating major characteristics of the disease in vitro, as well as to offer a powerful platform for pre-clinical drug development and for the testing of novel therapeutic options. In the present review, we summarize recent advances in iPSC-based disease modeling of mitochondrial cardiomyopathies and explore the patho-mechanistic insights as well as new therapeutic approaches that were uncovered with this experimental platform. Further, we discuss the challenges and limitations of this technology and provide an overview of the latest techniques to promote metabolic and functional maturation of iPSC-derived cardiomyocytes that might be necessary for modeling of mitochondrial disorders.
Collapse
Affiliation(s)
- Mario G Pavez-Giani
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| |
Collapse
|
23
|
Mittal A, Garg R, Bahl A, Khullar M. Molecular Mechanisms and Epigenetic Regulation in Diabetic Cardiomyopathy. Front Cardiovasc Med 2022; 8:725532. [PMID: 34977165 PMCID: PMC8716459 DOI: 10.3389/fcvm.2021.725532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus (DM) is an important lifestyle disease. Type 2 diabetes is one of the prime contributors to cardiovascular diseases (CVD) and diabetic cardiomyopathy (DbCM) and leads to increased morbidity and mortality in patients with DM. DbCM is a typical cardiac disease, characterized by cardiac remodeling in the presence of DM and in the absence of other comorbidities such as hypertension, valvular diseases, and coronary artery disease. DbCM is associated with defective cardiac metabolism, altered mitochondrial structure and function, and other physiological and pathophysiological signaling mechanisms such as oxidative stress, inflammation, myocardial apoptosis, and autophagy. Epigenetic modifiers are crucial players in the pathogenesis of DbCM. Thus, it is important to explore the role of epigenetic modifiers or modifications in regulating molecular pathways associated with DbCM. In this review, we have discussed the role of various epigenetic mechanisms such as histone modifications (acetylation and methylation), DNA methylation and non-coding RNAs in modulating molecular pathways involved in the pathophysiology of the DbCM.
Collapse
Affiliation(s)
- Anupam Mittal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajni Garg
- Council of Scientific and Industrial Research - Institute of Microbial Technology, Chandigarh, India
| | - Ajay Bahl
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
24
|
Zhao X, Liu S, Wang X, Chen Y, Pang P, Yang Q, Lin J, Deng S, Wu S, Fan G, Wang B. Diabetic cardiomyopathy: Clinical phenotype and practice. Front Endocrinol (Lausanne) 2022; 13:1032268. [PMID: 36568097 PMCID: PMC9767955 DOI: 10.3389/fendo.2022.1032268] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition of cardiac structure and function changes in diabetic patients without coronary artery disease, hypertension, and other types of heart diseases. DCM is not uncommon in people with diabetes, which increases the risk of heart failure. However, the treatment is scarce, and the prognosis is poor. Since 1972, one clinical study after another on DCM has been conducted. However, the complex phenotype of DCM still has not been fully revealed. This dilemma hinders the pace of understanding the essence of DCM and makes it difficult to carry out penetrating clinical or basic research. This review summarizes the literature on DCM over the last 40 years and discusses the overall perspective of DCM, phase of progression, potential clinical indicators, diagnostic and screening criteria, and related randomized controlled trials to understand DCM better.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Shengwang Liu
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Xiao Wang
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Yibing Chen
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Pai Pang
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Qianjing Yang
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Jingyi Lin
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Shuaishuai Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Shentao Wu
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Guanwei Fan
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Bin Wang
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| |
Collapse
|
25
|
Peng M, Liu H, Ji Q, Ma P, Niu Y, Ning S, Sun H, Pang X, Yang Y, Zhang Y, Han J, Hao G. Fufang Xueshuantong Improves Diabetic Cardiomyopathy by Regulating the Wnt/ β-Catenin Pathway. Int J Endocrinol 2022; 2022:3919161. [PMID: 36237833 PMCID: PMC9553353 DOI: 10.1155/2022/3919161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/02/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the main complications of diabetic patients and the major reason for the high prevalence of heart failure in diabetic patients. Fufang Xueshuantong (FXST) is a traditional Chinese medicine formula commonly used in the treatment of diabetic retinopathy and stable angina pectoris. However, the role of FXST in DCM has not yet been clarified. This study was conducted to investigate the effects of FXST on diabetic myocardial lesions and reveal its molecular mechanism. The rats were intraperitoneally injected with 65 mg/kg streptozotocin (STZ) to induce diabetes mellitus (DM). DM rats were given saline or FXST. The rats in the control group were intraperitoneally injected with an equal amount of sodium citrate buffer and gavaged with saline. After 12 weeks, echocardiography, heart weight index (HWI), and myocardial pathological changes were determined. The expression of transforming growth factor-beta1 (TGF-β1), collagen I, and collagen III was examined using immunofluorescence staining and western blot. The expressions of Wnt/β-catenin signaling pathway-related proteins and mRNA were detected by western blot and real-time PCR. The results showed that FXST significantly improved cardiac function, ameliorated histopathological changes, and decreased HWI in the DM rats. FXST significantly inhibited the expression of myocardial TGF-β1, collagen I, and collagen III in DM rats. Furthermore, FXST significantly inhibited the Wnt/β-catenin pathway. Taken together, FXST has a protective effect on DCM, which might be mediated by suppressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Meizhong Peng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hanying Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingxuan Ji
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pan Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiting Niu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shangqiu Ning
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huihui Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinxin Pang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuqian Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Zhang
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Han
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Gansu Provincial Hospital of Traditional Chinese Medicine, Gansu, China
| |
Collapse
|
26
|
Preethi KA, Selvakumar SC, Sekar D. Diagnostic and Therapeutic Application of Exosomal microRNAs Inducing Inflammation in Type 2 Diabetes Mellitus. Crit Rev Immunol 2022; 42:1-11. [PMID: 36374817 DOI: 10.1615/critrevimmunol.2022044927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Diabetes mellitus is a class of noncommunicable chronic metabolic disorders marked by hyperglycemia due to insulin production, insulin action or both and has reached epidemic levels around the world. The two most frequent types of diabetes are type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Despite substantial improvements in the knowledge and treatment of DM, the associated incidence and mortality rates remain steadily increased. Reliable markers for the early detection, monitoring and focused treatment of DM are desperately required. Conversely, microRNAs (miRNAs) have received much significance due to their regulatory involvement in gene expression. Fascinatingly, exosomes can be enclosed into miRNAs to transport or distribute them into the target cells or tissues in which they have a physiological regulatory action. Thus, exosomal miRNAs are proving to be important regulators in the establishment and maintenance of DM, however, further mode of action will be needed to investigate in order to fully comprehend the pathophysiological process. Hereby, this review outlines the recent findings on the role of exosomal miRNAs intending to understand the precise function in diagnostic and therapeutic aspects in T2DM disease.
Collapse
Affiliation(s)
- K Auxzilia Preethi
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - Sushmaa Chandralekha Selvakumar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - Durairaj Sekar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| |
Collapse
|
27
|
Xuan C, Luo Y, Xiong Y, Zhang Y, Tao C, Cao W. Multitarget mechanism of Yiqi Jiedu Huayu decoction on diabetic cardiomyopathy based on network pharmacology. Eur J Integr Med 2021. [DOI: 10.1016/j.eujim.2021.101388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
28
|
Su JH, Luo MY, Liang N, Gong SX, Chen W, Huang WQ, Tian Y, Wang AP. Interleukin-6: A Novel Target for Cardio-Cerebrovascular Diseases. Front Pharmacol 2021; 12:745061. [PMID: 34504432 PMCID: PMC8421530 DOI: 10.3389/fphar.2021.745061] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Cardio-Cerebrovascular Disease is a collective term for cardiovascular disease and cerebrovascular disease, being a serious threat to human health. A growing number of studies have proved that the content of inflammatory factors or mediators determines the stability of vascular plaque and the incidence of cardio-cerebrovascular event, and involves in the process of Cardio-Cerebrovascular Diseases. Interleukin-6 is a widely used cytokine that causes inflammation and oxidative stress, which would further result in cardiac and cerebral injury. The increased expression of interleukin-6 is closely related to atherosclerosis, myocardial infarction, heart failure and ischemic stroke. It is a key risk factor for these diseases by triggering inflammatory reaction and inducing other molecules release. Therefore, interleukin-6 may become a potential target for Cardio-Cerebrovascular Diseases in the future. This paper is aimed to discuss the expression changes and pathological mechanisms of interleukin-6 in Cardio-Cerebrovascular Diseases, and to provide a novel strategy for the prevention and treatment of Cardio-Cerebrovascular Diseases.
Collapse
Affiliation(s)
- Jian-Hui Su
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Meng-Yi Luo
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Na- Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Chen
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
29
|
Salvatore T, Pafundi PC, Galiero R, Albanese G, Di Martino A, Caturano A, Vetrano E, Rinaldi L, Sasso FC. The Diabetic Cardiomyopathy: The Contributing Pathophysiological Mechanisms. Front Med (Lausanne) 2021; 8:695792. [PMID: 34277669 PMCID: PMC8279779 DOI: 10.3389/fmed.2021.695792] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals with diabetes mellitus (DM) disclose a higher incidence and a poorer prognosis of heart failure (HF) than non-diabetic people, even in the absence of other HF risk factors. The adverse impact of diabetes on HF likely reflects an underlying “diabetic cardiomyopathy” (DM–CMP), which may by exacerbated by left ventricular hypertrophy and coronary artery disease (CAD). The pathogenesis of DM-CMP has been a hot topic of research since its first description and is still under active investigation, as a complex interplay among multiple mechanisms may play a role at systemic, myocardial, and cellular/molecular levels. Among these, metabolic abnormalities such as lipotoxicity and glucotoxicity, mitochondrial damage and dysfunction, oxidative stress, abnormal calcium signaling, inflammation, epigenetic factors, and others. These disturbances predispose the diabetic heart to extracellular remodeling and hypertrophy, thus leading to left ventricular diastolic and systolic dysfunction. This Review aims to outline the major pathophysiological changes and the underlying mechanisms leading to myocardial remodeling and cardiac functional derangement in DM-CMP.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
30
|
Cellular mechanisms and recommended drug-based therapeutic options in diabetic cardiomyopathy. Pharmacol Ther 2021; 228:107920. [PMID: 34171330 DOI: 10.1016/j.pharmthera.2021.107920] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus (DM) is associated with a specific cardiac phenotype characterized by structural and functional alterations. This so-called diabetic cardiomyopathy (DM CM) is clinically relevant as patients with DM show high incidence of heart failure. Mechanistically, several parameters interact on the cardiomyocyte level leading to increased inflammation, apoptosis, reactive oxygen species and altered calcium signaling. This in turn provokes functional myocardial changes that might inter alia play into the worsened clinical outcome in DM patients. Therefore, efficient therapeutic options are urgently needed. This review focuses on mechanistic effects of currently recommended antidiabetic treatment and heart failure therapy for DM CM.
Collapse
|
31
|
Kumric M, Ticinovic Kurir T, Borovac JA, Bozic J. Role of novel biomarkers in diabetic cardiomyopathy. World J Diabetes 2021; 12:685-705. [PMID: 34168722 PMCID: PMC8192249 DOI: 10.4239/wjd.v12.i6.685] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/22/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is commonly defined as cardiomyopathy in patients with diabetes mellitus in the absence of coronary artery disease and hypertension. As DCM is now recognized as a cause of substantial morbidity and mortality among patients with diabetes mellitus and clinical diagnosis is still inappropriate, various expert groups struggled to identify a suitable biomarker that will help in the recognition and management of DCM, with little success so far. Hence, we thought it important to address the role of biomarkers that have shown potential in either human or animal studies and which could eventually result in mitigating the poor outcomes of DCM. Among the array of biomarkers we thoroughly analyzed, long noncoding ribonucleic acids, soluble form of suppression of tumorigenicity 2 and galectin-3 seem to be most beneficial for DCM detection, as their plasma/serum levels accurately correlate with the early stages of DCM. The combination of relatively inexpensive and accurate speckle tracking echocardiography with some of the highlighted biomarkers may be a promising screening method for newly diagnosed diabetes mellitus type 2 patients. The purpose of the screening test would be to direct affected patients to more specific confirmation tests. This perspective is in concordance with current guidelines that accentuate the importance of an interdisciplinary team-based approach.
Collapse
Affiliation(s)
- Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
| | - Tina Ticinovic Kurir
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
- Department of Endocrinology, University Hospital of Split, Split 21000, Croatia
| | - Josip A Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
- Emergency Medicine, Institute of Emergency Medicine of Split-Dalmatia County, Split 21000, Croatia
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
| |
Collapse
|
32
|
Abukhalil MH, Althunibat OY, Aladaileh SH, Al-Amarat W, Obeidat HM, Al-Khawalde AAMA, Hussein OE, Alfwuaires MA, Algefare AI, Alanazi KM, Al-Swailmi FK, Arab HH, Mahmoud AM. Galangin attenuates diabetic cardiomyopathy through modulating oxidative stress, inflammation and apoptosis in rats. Biomed Pharmacother 2021; 138:111410. [PMID: 33752930 DOI: 10.1016/j.biopha.2021.111410] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/14/2021] [Accepted: 02/16/2021] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular complications are the leading cause of morbidity in diabetes. Oxidative stress and inflammation are implicated in the development and progression of diabetic cardiomyopathy (DCM). This study explored the cardioprotective effect of galangin (Gal), a natural flavonoid with radical-scavenging and anti-inflammatory activities, in diabetic rats. An experimental diabetic rat model was achieved by a single injection of 50 mg/kg streptozotocin. Gal (15 mg/kg) was administered daily for six weeks and the samples were then collected. Diabetic rats exhibited hyperglycemia, increased glycosylated hemoglobin, triglycerides and cholesterol levels and reduced serum insulin. Serum troponin I, CK-MB and LDH were increased in diabetic rats. Furthermore, hearts of diabetic rats were characterized by elevated malondialdehyde, protein carbonyl, NF-κB p65, TNF-α, IL-1β, iNOS, IL-6, Bax, caspase-3 and 8-Oxo-dG, and decreased superoxide dismutase, catalase, reduced GSH, and Bcl-2. Gal ameliorated hyperglycemia, dyslipidemia, and heart function markers, and prevented histopathological alterations in diabetic rats. In addition, Gal attenuated cardiac oxidative injury, inflammation and apoptosis, and boosted antioxidant defenses. In conclusion, Gal has a protective effect on cardiomyopathy by attenuating hyperglycemia, dyslipidemia, oxidative stress and inflammation in diabetic rats.
Collapse
Affiliation(s)
- Mohammad H Abukhalil
- Department of Biology, Faculty of Science, Al-Hussein Bin Talal University, Ma'an 71111, Jordan; Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an 71111, Jordan.
| | - Osama Y Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an 71111, Jordan
| | - Saleem H Aladaileh
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an 71111, Jordan; Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Wesam Al-Amarat
- Department of Medical Support, Al-karak University College, Al-Balqa' Applied University, As-Salt 206, Jordan
| | - Heba M Obeidat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an 71111, Jordan
| | - Alayn' Al-Marddyah A Al-Khawalde
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an 71111, Jordan
| | - Omnia E Hussein
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Manal A Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Abdulmohsen I Algefare
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Khalid M Alanazi
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Farhan K Al-Swailmi
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ayman M Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt; Biotechnology Department, Research Institute of Medicinal and Aromatic Plants, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
33
|
A Novel ALDH2 Activator AD-9308 Improves Diastolic and Systolic Myocardial Functions in Streptozotocin-Induced Diabetic Mice. Antioxidants (Basel) 2021; 10:antiox10030450. [PMID: 33805825 PMCID: PMC7998151 DOI: 10.3390/antiox10030450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus has reached epidemic proportion worldwide. One of the diabetic complications is cardiomyopathy, characterized by early left ventricular (LV) diastolic dysfunction, followed by development of systolic dysfunction and ventricular dilation at a late stage. The pathogenesis is multifactorial, and there is no effective treatment yet. In recent years, 4-hydroxy-2-nonenal (4-HNE), a toxic aldehyde generated from lipid peroxidation, is implicated in the pathogenesis of cardiovascular diseases. Its high bioreactivity toward proteins results in cellular damage. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is the major enzyme that detoxifies 4-HNE. The development of small-molecule ALDH2 activator provides an opportunity for treating diabetic cardiomyopathy. This study found that AD-9308, a water-soluble andhighly selective ALDH2 activator, can improve LV diastolic and systolic functions, and wall remodeling in streptozotocin-induced diabetic mice. AD-9308 treatment dose-dependently lowered serum 4-HNE levels and 4-HNE protein adducts in cardiac tissue from diabetic mice, accompanied with ameliorated myocardial fibrosis, inflammation, and apoptosis. Improvements of mitochondrial functions, sarco/endoplasmic reticulumcalcium handling and autophagy regulation were also observed in diabetic mice with AD-9308 treatment. In conclusion, ADLH2 activation effectively ameliorated diabetic cardiomyopathy, which may be mediated through detoxification of 4-HNE. Our findings highlighted the therapeutic potential of ALDH2 activation for treating diabetic cardiomyopathy.
Collapse
|
34
|
Ifuku M, Takahashi K, Hosono Y, Iso T, Ishikawa A, Haruna H, Takubo N, Komiya K, Kurita M, Ikeda F, Watada H, Shimizu T. Left atrial dysfunction and stiffness in pediatric and adult patients with Type 1 diabetes mellitus assessed with speckle tracking echocardiography. Pediatr Diabetes 2021; 22:303-319. [PMID: 33094524 DOI: 10.1111/pedi.13141] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/05/2020] [Accepted: 10/15/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Subclinical diastolic dysfunction in patients with Type 1 diabetes mellitus (T1DM) caused by myocardial injury due to diabetic cardiomyopathy leads to a high risk of death and heart failure. This myocardial injury extends not only to the left ventricle (LV) but also to the left atrium (LA). However, LA function in children and young adults with T1DM has not been extensively studied. OBJECTIVE Therefore, the aim of this study was to assess LA dysfunction in pediatric and adult patients with T1DM using LA strain analysis with echocardiography. SUBJECTS Fifty-three patients (median age: 23 [range: 5-41] years) with T1DM. METHODS We divided the patients into three age groups (D1: 5-14 years, D2: 15-24 years, D3: 25-41 years); 53 age- and sex-matched controls were divided into three corresponding groups (C1, C2, and C3). LA and LV functions were evaluated using echocardiography. RESULTS LA reservoir strain was lower in the D2 and D3 groups than in the C2 and C3 groups (P = 0.001, P = 0.004, respectively). LA conduit strain was lower in the D2 group than in the C2 group (P = 0.002). LA stiffness was significantly greater in the D3 group than in the C3 group (P < 0.001). CONCLUSIONS In patients with T1DM, LA phasic function decreased in adolescents and young adults, and LA stiffness increased in adult patients aged >30 years. LA phasic function and LA stiffness can be potentially used as early markers for diastolic dysfunction.
Collapse
Affiliation(s)
- Mayumi Ifuku
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Ken Takahashi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yu Hosono
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Takeshi Iso
- Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Akimi Ishikawa
- Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hidenori Haruna
- Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Noriyuki Takubo
- Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Kouji Komiya
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Mika Kurita
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Fuki Ikeda
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan.,Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
35
|
Grover A, Sharma K, Gautam S, Gautam S, Gulati M, Singh SK. Diabetes and Its Complications: Therapies Available, Anticipated and Aspired. Curr Diabetes Rev 2021; 17:397-420. [PMID: 33143627 DOI: 10.2174/1573399816666201103144231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/26/2020] [Accepted: 09/12/2020] [Indexed: 11/22/2022]
Abstract
Worldwide, diabetes ranks among the ten leading causes of mortality. Prevalence of diabetes is growing rapidly in low and middle income countries. It is a progressive disease leading to serious co-morbidities, which results in increased cost of treatment and over-all health system of the country. Pathophysiological alterations in Type 2 Diabetes (T2D) progressed from a simple disturbance in the functioning of the pancreas to triumvirate to ominous octet to egregious eleven to dirty dozen model. Due to complex interplay of multiple hormones in T2D, there may be multifaceted approach in its management. The 'long-term secondary complications' in uncontrolled diabetes may affect almost every organ of the body, and finally may lead to multi-organ dysfunction. Available therapies are inconsistent in maintaining long term glycemic control and their long term use may be associated with adverse effects. There is need for newer drugs, not only for glycemic control but also for prevention or mitigation of secondary microvascular and macrovascular complications. Increased knowledge of the pathophysiology of diabetes has contributed to the development of novel treatments. Several new agents like Glucagon Like Peptide - 1 (GLP-1) agonists, Dipeptidyl Peptidase IV (DPP-4) inhibitors, amylin analogues, Sodium-Glucose transport -2 (SGLT- 2) inhibitors and dual Peroxisome Proliferator-Activated Receptor (PPAR) agonists are available or will be available soon, thus extending the range of therapy for T2D, thereby preventing its long term complications. The article discusses the pathophysiology of diabetes along with its comorbidities, with a focus on existing and novel upcoming antidiabetic drugs which are under investigation. It also dives deep to deliberate upon the novel therapies that are in various stages of development. Adding new options with new mechanisms of action to the treatment armamentarium of diabetes may eventually help improve outcomes and reduce its economic burden.
Collapse
Affiliation(s)
- Anu Grover
- Ipca Laboratories, Mumbai - 400063, India
| | - Komal Sharma
- Bhupal Nobles' Institute of Pharmaceutical Sciences, Udaipur, India
| | - Suresh Gautam
- Department of Biochemistry, Pacific Institute of Medical Sciences, Udaipur, India
| | - Srishti Gautam
- Ravinder Nath Tagore Medical College and Maharana Bhupal Govt. Hospital, Udaipur, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab- 144411, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab- 144411, India
| |
Collapse
|
36
|
Zhang X, Hao Y. Beneficial Effects of Echinacoside on Diabetic Cardiomyopathy in Diabetic Db/Db Mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5575-5587. [PMID: 33376302 PMCID: PMC7755380 DOI: 10.2147/dddt.s276972] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/10/2020] [Indexed: 12/31/2022]
Abstract
Purpose In this study, we investigated the protective effects and mechanism of action of echinacoside (ECH) from cistanche tubulosa extract in cardiomyocytes of db/db diabetic mice. Methods Twenty healthy male db/db mice aged 8 weeks were randomly divided into db/db+ECH (n=10, ECH, 300 mg/(kg/d)), db/db (n=10, saline), and db/m control groups (n=9). Mice were monitored weekly for diet and activity. Mice were injected with 2% of pentobarbital sodium in week 10 and executed. Weight and free blood glucose (FBG) were measured weekly. Echocardiographs were used to detect cardiac function. HE staining, Sudan II staining, Masson’s trichrome staining and Tunel assays were used to evaluate myocardial tissue pathological changes, collagen fiber deposition, lipid accumulation and apoptosis rates in cardiomyocytes, respectively. Western blot and RT-PCR analysis were used to detect the expression of components of the PPAR-α/M-CPT-1 and p53/p38MAPK signaling axis. Results Compared to db/db mice, ECH groups showed lower blood glucose and lipid levels. Deterioration in cardiac function was also delayed following ECH treatment. Histopathological analysis showed that ECH significantly improved myocardial tissue in db/db mice, including reduced intercellular spaces, regular arrangements, improved extracellular matrix deposition, and reduced lipid accumulation. ECH also significantly reduced oxidative stress levels in myocardial tissue in db/db mice. Moreover, ECH inhibited PPAR-α/M-CPT-1 signaling, downregulated CD36, and upregulated glucose transporter type 4 (GLUT-4) expression in db/db mouse models of DCM. ECH also inhibited p53/p38MAPK signaling, downregulated caspase-3 and caspase-8, and upregulated Bcl-2/Bax in db/db mouse models of DCM. Conclusion ECH displays protective effects in DCM, including the inhibition of cardiac apoptosis and oxidative stress, and improved lipid metabolism in cardiomyocytes. ECH also inhibits cardiac apoptosis through its regulation of p53/p38MAPK signaling, and prevents lipid accumulation through suppression of the PPAR-α/M-CPT-1 signaling axis.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei, People's Republic of China
| | - Yarong Hao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei, People's Republic of China
| |
Collapse
|
37
|
Gopal K, Chahade JJ, Kim R, Ussher JR. The Impact of Antidiabetic Therapies on Diastolic Dysfunction and Diabetic Cardiomyopathy. Front Physiol 2020; 11:603247. [PMID: 33364978 PMCID: PMC7750477 DOI: 10.3389/fphys.2020.603247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic cardiomyopathy is more prevalent in people with type 2 diabetes mellitus (T2DM) than previously recognized, while often being characterized by diastolic dysfunction in the absence of systolic dysfunction. This likely contributes to why heart failure with preserved ejection fraction is enriched in people with T2DM vs. heart failure with reduced ejection fraction. Due to revised mandates from major health regulatory agencies, all therapies being developed for the treatment of T2DM must now undergo rigorous assessment of their cardiovascular risk profiles prior to approval. As such, we now have data from tens of thousands of subjects with T2DM demonstrating the impact of major therapies including the sodium-glucose co-transporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 receptor (GLP-1R) agonists, and dipeptidyl peptidase 4 (DPP-4) inhibitors on cardiovascular outcomes. Evidence to date suggests that both SGLT2 inhibitors and GLP-1R agonists improve cardiovascular outcomes, whereas DPP-4 inhibitors appear to be cardiovascular neutral, though evidence is lacking to determine the overall utility of these therapies on diastolic dysfunction or diabetic cardiomyopathy in subjects with T2DM. We herein will review the overall impact SLGT2 inhibitors, GLP-1R agonists, and DPP-4 inhibitors have on major parameters of diastolic function, while also highlighting the potential mechanisms of action responsible. A more complete understanding of how these therapies influence diastolic dysfunction will undoubtedly play a major role in how we manage cardiovascular disease in subjects with T2DM.
Collapse
Affiliation(s)
- Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Jadin J Chahade
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Ryekjang Kim
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
38
|
Haye A, Ansari MA, Rahman SO, Shamsi Y, Ahmed D, Sharma M. Role of AMP-activated protein kinase on cardio-metabolic abnormalities in the development of diabetic cardiomyopathy: A molecular landscape. Eur J Pharmacol 2020; 888:173376. [PMID: 32810493 DOI: 10.1016/j.ejphar.2020.173376] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Cardiovascular complications associated with diabetes mellitus remains a leading cause of morbidity and mortality across the world. Diabetic cardiomyopathy is a descriptive pathology that in absence of co-morbidities such as hypertension, dyslipidemia initially characterized by cardiac stiffness, myocardial fibrosis, ventricular hypertrophy, and remodeling. These abnormalities further contribute to diastolic dysfunctions followed by systolic dysfunctions and eventually results in clinical heart failure (HF). The clinical outcomes associated with HF are considerably worse in patients with diabetes. The complexity of the pathogenesis and clinical features of diabetic cardiomyopathy raises serious questions in developing a therapeutic strategy to manage cardio-metabolic abnormalities. Despite extensive research in the past decade the compelling approaches to manage and treat diabetic cardiomyopathy are limited. AMP-Activated Protein Kinase (AMPK), a serine-threonine kinase, often referred to as cellular "metabolic master switch". During the development and progression of diabetic cardiomyopathy, a plethora of evidence demonstrate the beneficial role of AMPK on cardio-metabolic abnormalities including altered substrate utilization, impaired cardiac insulin metabolic signaling, mitochondrial dysfunction and oxidative stress, myocardial inflammation, increased accumulation of advanced glycation end-products, impaired cardiac calcium handling, maladaptive activation of the renin-angiotensin-aldosterone system, endoplasmic reticulum stress, myocardial fibrosis, ventricular hypertrophy, cardiac apoptosis, and impaired autophagy. Therefore, in this review, we have summarized the findings from pre-clinical and clinical studies and provided a collective overview of the pathophysiological mechanism and the regulatory role of AMPK on cardio-metabolic abnormalities during the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Abdul Haye
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yasmeen Shamsi
- Department of Moalejat, School of Unani Medical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Danish Ahmed
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture Technology and Sciences, Allahabad, Uttar Pradesh, India
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
39
|
Mu J, Zhang D, Tian Y, Xie Z, Zou MH. BRD4 inhibition by JQ1 prevents high-fat diet-induced diabetic cardiomyopathy by activating PINK1/Parkin-mediated mitophagy in vivo. J Mol Cell Cardiol 2020; 149:1-14. [PMID: 32941882 DOI: 10.1016/j.yjmcc.2020.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
BRD4 is a member of the BET family of epigenetic regulators. Inhibition of BRD4 by the selective bromodomain inhibitor JQ1, alleviates thoracic aortic constriction-induced cardiac hypertrophy and heart failure. However, whether BRD4 inhibition by JQ1 has therapeutic effect on diabetic cardiomyopathy, a major cause of heart failure in patients with Type 2 diabetes, remains unknown. Here, we discover a novel link between BRD4 and PINK1/Parkin-mediated mitophagy during diabetic cardiomyopathy. Upregulation of BRD4 in diabetic mouse hearts inhibits PINK1/Parkin-mediated mitophagy, resulting in accumulation of damaged mitochondria and subsequent impairment of cardiac structure and function. BRD4 inhibition by JQ1 improves mitochondrial function, and repairs the cardiac structure and function of the diabetic heart. These effects depended on rewiring of the BRD4-driven transcription and repression of PINK1. Deletion of Pink1 suppresses mitophagy, exacerbates cardiomyopathy, and abrogates the therapeutic effect of JQ1 on diabetic cardiomyopathy. Our results illustrate a valid therapeutic strategy for treating diabetic cardiomyopathy by inhibition of BRD4.
Collapse
Affiliation(s)
- Jing Mu
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, United States of America; Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, United States of America
| | - Donghong Zhang
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, United States of America
| | - Yunli Tian
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, United States of America
| | - Zhonglin Xie
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, United States of America.
| | - Ming-Hui Zou
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, United States of America; Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, United States of America.
| |
Collapse
|
40
|
Zhang Y, Qian H, Wu B, You S, Wu S, Lu S, Wang P, Cao L, Zhang N, Sun Y. E3 Ubiquitin ligase NEDD4 family‑regulatory network in cardiovascular disease. Int J Biol Sci 2020; 16:2727-2740. [PMID: 33110392 PMCID: PMC7586430 DOI: 10.7150/ijbs.48437] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Protein ubiquitination represents a critical modification occurring after translation. E3 ligase catalyzes the covalent binding of ubiquitin to the protein substrate, which could be degraded. Ubiquitination as an important protein post-translational modification is closely related to cardiovascular disease. The NEDD4 family, belonging to HECT class of E3 ubiquitin ligases can recognize different substrate proteins, including PTEN, ENaC, Nav1.5, SMAD2, PARP1, Septin4, ALK1, SERCA2a, TGFβR3 and so on, via the WW domain to catalyze ubiquitination, thus participating in multiple cardiovascular-related disease such as hypertension, arrhythmia, myocardial infarction, heart failure, cardiotoxicity, cardiac hypertrophy, myocardial fibrosis, cardiac remodeling, atherosclerosis, pulmonary hypertension and heart valve disease. However, there is currently no review comprehensively clarifying the important role of NEDD4 family proteins in the cardiovascular system. Therefore, the present review summarized recent studies about NEDD4 family members in cardiovascular disease, providing novel insights into the prevention and treatment of cardiovascular disease. In addition, assessing transgenic animals and performing gene silencing would further identify the ubiquitination targets of NEDD4. NEDD4 quantification in clinical samples would also constitute an important method for determining NEDD4 significance in cardiovascular disease.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Pingyuan Wang
- Staff scientist, Center for Molecular Medicine National Heart Lung and Blood Institute, National Institutes of Health, the United States
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
41
|
Yuan H, Xu J, Zhu Y, Li L, Wang Q, Yu Y, Zhou B, Liu Y, Xu X, Wang Z. Activation of calcium‑sensing receptor‑mediated autophagy in high glucose‑induced cardiac fibrosis in vitro. Mol Med Rep 2020; 22:2021-2031. [PMID: 32705187 PMCID: PMC7411369 DOI: 10.3892/mmr.2020.11277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 05/28/2020] [Indexed: 12/28/2022] Open
Abstract
Myocardial fibrosis is a major complication of diabetic cardiomyopathy (DCM) that is primarily caused by cardiac fibroblasts that are highly activated by persistent hyperglycemic stimulation, resulting in excessive collagen deposition. Calcium sensing receptor (CaSR) is a member of the G protein-coupled receptor superfamily and regulates intracellular calcium concentrations, which are associated with numerous diseases, including myocardial infarction, tumors and pulmonary hypertension. However, whether CaSR participates in the pathological process of myocardial fibrosis in DCM remains unknown. The present study aimed to investigate the mechanism via which CaSR regulates high glucose (HG)-induced cardiac fibrosis in vitro. HG treated-cardiac fibroblast (CFs) were used and western blotting, immunoprecipitation, Cell Counting Kit-8 assay, ELISA and transfection technology were performed to examine the role of CaSR. In the HG group, treatment with HG increased CaSR, α-smooth muscle actin, collagen I/III and matrix metalloproteinase 2/9 expression and enhanced autophagosome generation and CF proliferation. Furthermore, CaSR activation upregulated the expression of Smad ubiquitin regulatory factor 2 (Smurf2), which led to increased intracellular Ca2+ concentrations, increased ubiquitination levels of SKI like proto-oncogene and Smad7 and autophagy activation. Furthermore, the CaSR agonist (R568) or the CaSR inhibitor (Calhex231) and Smurf2-small interfering RNA promoted or inhibited HG-induced alterations, including the enhanced and weakened effects, respectively. Taken together, the results from the present study suggested that increased CaSR expression in CFs activated the Smurf2-ubiquitin proteasome and autophagy, causing excessive CF proliferation and extensive collagen deposition, which resulted in HG-induced myocardial fibrosis. These findings indicated a novel pathogenesis of DCM and may provide a novel strategy for the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Medical Functional Experiment and Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jiyu Xu
- Department of Medical Functional Experiment and Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yanfei Zhu
- Department of Medical Functional Experiment and Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Li Li
- Department of Medical Functional Experiment and Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Qi Wang
- Department of General Surgery, Mudanjiang First People's Hospital, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yaquan Yu
- Department of Gastroenterology, Yang Zhou Hong Quan Hospital, Yangzhou, Jiangsu 225000, P.R. China
| | - Bin Zhou
- Department of Endocrinology, Mudanjiang Cardiovascular Hospital, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yi Liu
- Department of Medical Functional Experiment and Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xiaoyi Xu
- Department of Medical Functional Experiment and Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Zhilong Wang
- Department of Postgraduate Management, The First Clinical Medicine School, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
42
|
Carracedo J, Alique M, Ramírez-Carracedo R, Bodega G, Ramírez R. Endothelial Extracellular Vesicles Produced by Senescent Cells: Pathophysiological Role in the Cardiovascular Disease Associated with all Types of Diabetes Mellitus. Curr Vasc Pharmacol 2020; 17:447-454. [PMID: 30124156 DOI: 10.2174/1570161116666180820115726] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022]
Abstract
Endothelial senescence-associated with aging or induced prematurely in pathological situations, such as diabetes, is a first step in the development of Cardiovascular Disease (CVDs) and particularly inflammatory cardiovascular diseases. The main mechanism that links endothelial senescence and the progression of CVDs is the production of altered Extracellular Vesicles (EVs) by senescent endothelial cells among them, Microvesicles (MVs). MVs are recognized as intercellular signaling elements that play a key role in regulating tissue homeostasis. However, MVs produced by damage cell conveyed epigenetic signals, mainly involving microRNAs, which induce many of the injured responses in other vascular cells leading to the development of CVDs. Many studies strongly support that the quantification and characterization of the MVs released by senescent endothelial cells may be useful diagnostic tools in patients with CVDs, as well as a future therapeutic target for these diseases. In this review, we summarize the current knowledge linking senescence-associated MVs to the development of CVDs and discuss the roles of these MVs, in particular, in diabetic-associated increases the risk of CVDs.
Collapse
Affiliation(s)
- Julia Carracedo
- Department of Genetic, Physiology and Microbiology, Faculty of Biology, Complutense University/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Matilde Alique
- Biology Systems Department, Physiology, Alcala University, Alcala de Henares, Madrid, Spain
| | - Rafael Ramírez-Carracedo
- Cardiovascular Joint Research Unit, University Francisco de Vitoria/ University Hospital Ramon y Cajal Research Unit (IRYCIS), Madrid, Spain
| | - Guillermo Bodega
- Biomedicine and Biotechnology Department, Alcala University, Alcala de Henares, Madrid, Spain
| | - Rafael Ramírez
- Biology Systems Department, Physiology, Alcala University, Alcala de Henares, Madrid, Spain
| |
Collapse
|
43
|
Roberti SL, Higa R, Sato H, Gomez Ribot D, Capobianco E, Jawerbaum A. Olive oil supplementation prevents extracellular matrix deposition and reduces prooxidant markers and apoptosis in the offspring´s heart of diabetic rats. Reprod Toxicol 2020; 95:137-147. [PMID: 32417168 DOI: 10.1016/j.reprotox.2020.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022]
Abstract
Maternal diabetes induces fetal programming of cardiovascular diseases. Diabetes induced-cardiac fibrosis is a process that may start in utero and may be related to the prooxidant/proinflammatory environment. The aim of this study was to investigate the effect of a maternal diet enriched in olive oil on the levels of components and regulators of the extracellular matrix, on prooxidant markers and on apoptosis rate in the heart of 21-day-old offspring of diabetic rats. Maternal diabetes was induced by neonatal administration of streptozotocin. During pregnancy, diabetic and control rats were fed with diets supplemented or not with 6% olive oil. The heart of the offspring was studied at 21 days of age. We found increased deposition of collagen IV and fibronectin in the offspring´s heart of diabetic rats, which was prevented by the maternal diets enriched in olive oil. Increases in connective tissue growth factor were also prevented by the maternal diets enriched in olive oil. Prooxidant markers as well as apoptosis, which were increased in the heart of the offspring of diabetic rats, were prevented by the maternal olive oil dietary treatment. Our findings identified powerful effects of a maternal diet enriched in olive oil on the prevention of increased extracellular matrix deposition and increased prooxidant markers in the heart of 21-day-old offspring of diabetic rats.
Collapse
Affiliation(s)
- Sabrina L Roberti
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Romina Higa
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Hugo Sato
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Dalmiro Gomez Ribot
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Evangelina Capobianco
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina.
| |
Collapse
|
44
|
Hu J, Lu X, Zhang X, Shao X, Wang Y, Chen J, Zhao B, Li S, Xu C, Wei C. Exogenous spermine attenuates myocardial fibrosis in diabetic cardiomyopathy by inhibiting endoplasmic reticulum stress and the canonical Wnt signaling pathway. Cell Biol Int 2020; 44:1660-1670. [PMID: 32304136 DOI: 10.1002/cbin.11360] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/30/2020] [Accepted: 04/11/2020] [Indexed: 12/13/2022]
Abstract
Myocardial fibrosis is one of the main pathological manifestations of diabetic cardiomyopathy (DCM). Spermine (SPM), a product of polyamine metabolism, plays an important role in many cardiac diseases including hypertrophy, ischemia, and infarction, but its role in diabetic myocardial fibrosis has not been clarified. This study aimed to investigate the role of polyamine metabolism, specifically SPM, in diabetic myocardial fibrosis and to explore the related mechanisms. We used intraperitoneal injection of streptozotocin (STZ, 60 mg/kg) in Wistar rats and high glucose (HG, 40 mM) stimulated cardiac fibroblasts (CFs) to established a type 1 diabetes (T1D) model in vivo and in vitro, which were pretreated with exogenous SPM (5 mg/kg per day and 5 μM). The results showed that hyperglycemia induced the expression of the key polyamine synthesis enzyme ornithine decarboxylase (ODC) decreased and the key catabolic enzyme spermidine/spermine N1 -acetyltransferase (SSAT) increased compared with those in the control group. The body weight, blood insulin level, and cardiac ejection function were decreased, while blood glucose, heart weight, the ratio of heart weight to body weight, myocardial interstitial collagen deposition, and endoplasmic reticulum stress (ERS)-related protein (glucose-regulated protein-78, glucose-regulated protein-94, activating transcription factor-4, and C/EBP homology protein) expression in the T1D group were all significantly increased. HG also caused an increased expression of Wnt3, β-catenin (in cytoplasm and nucleus), while Axin2 and phosphorylated β-catenin decreased. Exogenous SPM improved the above changes caused by polyamine metabolic disorders. In conclusion, polyamine metabolism disorder occurs in the myocardial tissue of diabetic rats, causing myocardial fibrosis and ERS. Exogenous SPM plays a myocardial protective role via inhibiting of ERS and the canonical Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jing Hu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xiaoxiao Lu
- Department of Physical Diagnostics, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Xinying Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xiaoting Shao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yuehong Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Junting Chen
- Department of Anesthesiology, Harbin Medical University Fourth Hospital, Harbin, China
| | - Bingbing Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Siwei Li
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Can Wei
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Yazdani F, Shahidi F, Karimi P. The effect of 8 weeks of high-intensity interval training and moderate-intensity continuous training on cardiac angiogenesis factor in diabetic male rats. J Physiol Biochem 2020; 76:291-299. [PMID: 32157499 DOI: 10.1007/s13105-020-00733-5] [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: 08/22/2019] [Accepted: 02/13/2020] [Indexed: 01/04/2023]
Abstract
The balance of pro-angiogenic and anti-angiogenic factors has a significant role in the development of diabetic cardiomyopathy. The purpose of this study was to examine the effect of 8 weeks of high-intensity interval training (HIIT) and moderate intensity continuous training (MICT) on the myocardial angiogenic factors and histological changes in male diabetic rats. Thirty-two male Wistar rats were randomly assigned into four groups: healthy non-exercised control, diabetic (D), D + HIIT, and D+ MICT groups. Diabetes type 2 was induced by a high-fat diet for 2 weeks and a single injection of streptozotocin. Following confirmation of diabetes, animals were subjected to HIIT (90 to 95% of VO2max) or MICT (50-65% of VO2max) protocols 5 days a week for 8 weeks. Western blotting was used for detection of protein expressions of vascular endothelial growth factor (VEGF), transforming growth factor-beta (TGF-β), matrix metalloproteinase-2 (MMP2), and tissue inhibitor of matrix metalloproteinase-2 (TIMP2) in the left ventricle. In addition, baseline and final blood glucose and body weight were measured. Histological changes were evaluated using H&E and Masson's trichrome staining. The results showed that exercise increased protein levels of pro-angiogenic factors while reduced anti-angiogenic factors protein levels in diabetic animals. These changes were followed by increased capillary density and reduced interstitial fibrosis in the left ventricle. Moreover, the MICT was superior to HIIT in enhancing angiogenic factors and attenuation of blood glucose and fibrosis in the diabetic rats. These findings confirm the effectiveness of exercise, particularly MICT, in the improvement of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Faramarz Yazdani
- Department of Physiology, Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Fereshteh Shahidi
- Department of Physiology, Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
46
|
Shang R, Lal N, Puri K, Hussein B, Rodrigues B. Involvement of Heparanase in Endothelial Cell-Cardiomyocyte Crosstalk. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:721-745. [PMID: 32274734 DOI: 10.1007/978-3-030-34521-1_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditionally, the management of diabetes has focused mainly on controlling high blood glucose levels. Unfortunately, despite valiant efforts to normalize this blood glucose, poor medication management predisposes these patients to heart failure. Following diabetes, how the heart utilizes different sources of fuel for energy is key to the development of heart failure. The diabetic heart switches from using both glucose and fats, to predominately using fats as an energy resource for maintaining its activities. This transformation to using fats as an exclusive source of energy is helpful in the initial stages of the disease and is tightly controlled. However, over the progression of diabetes, there is a loss of this controlled supply and use of fats, which ultimately has terrible consequences since the uncontrolled use of fats produces toxic by-products which weaken heart function and cause heart disease. Heparanase is a key player that directs how much fats are provided to the heart and does so in association with several partners like LPL and VEGFs. Together, they regulate the amount of fats supplied, and their subsequent breakdown to provide energy. Following diabetes, there is a disruption in this network resulting in fat oversupply and cell death. Understanding how the heparanase-LPL-VEGFs "ensemble" cooperates, and its dysfunction in the diabetic heart would be useful in restoring metabolic equilibrium and limiting diabetes-related cardiac damage.
Collapse
Affiliation(s)
- Rui Shang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Nathaniel Lal
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Karanjit Puri
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Bahira Hussein
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
47
|
Précoma DB, Oliveira GMMD, Simão AF, Dutra OP, Coelho OR, Izar MCDO, Póvoa RMDS, Giuliano IDCB, Alencar Filho ACD, Machado CA, Scherr C, Fonseca FAH, Santos Filho RDD, Carvalho TD, Avezum Á, Esporcatte R, Nascimento BR, Brasil DDP, Soares GP, Villela PB, Ferreira RM, Martins WDA, Sposito AC, Halpern B, Saraiva JFK, Carvalho LSF, Tambascia MA, Coelho-Filho OR, Bertolami A, Correa Filho H, Xavier HT, Faria-Neto JR, Bertolami MC, Giraldez VZR, Brandão AA, Feitosa ADDM, Amodeo C, Souza DDSMD, Barbosa ECD, Malachias MVB, Souza WKSBD, Costa FAAD, Rivera IR, Pellanda LC, Silva MAMD, Achutti AC, Langowiski AR, Lantieri CJB, Scholz JR, Ismael SMC, Ayoub JCA, Scala LCN, Neves MF, Jardim PCBV, Fuchs SCPC, Jardim TDSV, Moriguchi EH, Schneider JC, Assad MHV, Kaiser SE, Lottenberg AM, Magnoni CD, Miname MH, Lara RS, Herdy AH, Araújo CGSD, Milani M, Silva MMFD, Stein R, Lucchese FA, Nobre F, Griz HB, Magalhães LBNC, Borba MHED, Pontes MRN, Mourilhe-Rocha R. Updated Cardiovascular Prevention Guideline of the Brazilian Society of Cardiology - 2019. Arq Bras Cardiol 2019; 113:787-891. [PMID: 31691761 PMCID: PMC7020870 DOI: 10.5935/abc.20190204] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Affiliation(s)
- Dalton Bertolim Précoma
- Pontifícia Universidade Católica do Paraná (PUC-PR), Curitiba, PR - Brazil
- Sociedade Hospitalar Angelina Caron, Campina Grande do Sul, PR - Brazil
| | | | | | | | | | | | | | | | | | | | | | | | - Raul Dias Dos Santos Filho
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, SP - Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP - Brazil
| | - Tales de Carvalho
- Clínica Cardiosport de Prevenção e Reabilitação, Florianópolis, SC - Brazil
- Departamento de Ergometria e Reabilitação Cardiovascular da Sociedade Brazileira de Cardiologia (DERC/SBC), Rio de Janeiro, RJ - Brazil
- Universidade do Estado de Santa Catarina (UDESC), Florianópolis, SC - Brazil
| | - Álvaro Avezum
- Hospital Alemão Oswaldo Cruz, São Paulo, SP - Brazil
| | - Roberto Esporcatte
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ - Brazil
- Hospital Pró-Cardíaco, Rio de Janeiro, RJ - Brazil
| | - Bruno Ramos Nascimento
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, MG - Brazil
| | - David de Pádua Brasil
- Faculdade de Ciências Médicas de Minas Gerias (CMMG) da Fundação Educacional Lucas Machado (FELUMA), Belo Horizonte, MG - Brazil
- Hospital Universitário Ciências Médicas (HUCM), Belo Horizonte, MG - Brazil
- Universidade Federal de Lavas (UFLA), Lavras, MG - Brazil
| | - Gabriel Porto Soares
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ - Brazil
- Universidade de Vassouras, Vassouras, RJ - Brazil
| | - Paolo Blanco Villela
- Hospital Universitário Clementino Fraga Filho da Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ - Brazil
- Hospital Samaritano, Rio de Janeiro, RJ - Brazil
| | | | - Wolney de Andrade Martins
- Universidade Federal Fluminense (UFF), Niterói, RJ - Brazil
- Complexo Hospitalar de Niterói, Niterói, RJ - Brazil
| | - Andrei C Sposito
- Universidade Estadual de Campinas (UNICAMP), Campina, SP - Brazil
| | - Bruno Halpern
- Universidade de São Paulo (USP), São Paulo, SP - Brazil
| | | | | | | | | | | | | | | | | | | | - Viviane Zorzanelli Rocha Giraldez
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, SP - Brazil
| | | | | | - Celso Amodeo
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP - Brazil
| | | | | | | | | | | | | | - Lucia Campos Pellanda
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS - Brazil
- Fundação Universitária de cardiologia do RS (ICFUC), Porto Alegre, RS - Brazil
| | | | | | | | | | - Jaqueline Ribeiro Scholz
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, SP - Brazil
| | | | - José Carlos Aidar Ayoub
- Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP - Brazil
- Instituto de Moléstias Cardiovasculares, São José do Rio Preto, SP - Brazil
| | | | - Mario Fritsch Neves
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ - Brazil
| | | | | | | | | | - Jamil Cherem Schneider
- SOS Cardio, Florianópolis, SC - Brazil
- Universidade do Sul de SC (Unisul), Florianópolis, SC - Brazil
| | | | | | - Ana Maria Lottenberg
- Hospital Israelita Albert Einstein, São Paulo, SP - Brazil
- Laboratório de Lípides (LIM10), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, São Paulo, SP - Brazil
| | | | - Marcio Hiroshi Miname
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, SP - Brazil
| | - Roberta Soares Lara
- Instituto de Nutrição Roberta Lara, Itu, SP - Brazil
- Diadia Nutrição e Gastronomia, Itu, SP - Brazil
| | - Artur Haddad Herdy
- Instituto de Cardiologia de Santa Catarina, São José, SC - Brazil
- Clínica Cardiosport de Prevenção e Reabilitação, Florianópolis, SC - Brazil
| | | | | | | | - Ricardo Stein
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brazil
| | | | | | - Hermilo Borba Griz
- Hospital Santa Joana Recife, Recife, PE - Brazil
- Hospital Agamenon Magalhães, Recife, PE - Brazil
| | | | | | - Mauro Ricardo Nunes Pontes
- Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, RS - Brazil
- Hospital São Francisco, Porto Alegre, RS - Brazil
| | - Ricardo Mourilhe-Rocha
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ - Brazil
- Hospital Pró-Cardíaco, Rio de Janeiro, RJ - Brazil
| |
Collapse
|
48
|
Andrographolide Protects against HG-Induced Inflammation, Apoptosis, Migration, and Impairment of Angiogenesis via PI3K/AKT-eNOS Signalling in HUVECs. Mediators Inflamm 2019; 2019:6168340. [PMID: 31686985 PMCID: PMC6800917 DOI: 10.1155/2019/6168340] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/01/2019] [Accepted: 05/08/2019] [Indexed: 12/22/2022] Open
Abstract
Andrographolide (Andr) is a major component isolated from the plant Andrographis paniculata. Inflammation, apoptosis, and impaired angiogenesis are implicated in the pathogenesis of high glucose (HG)-induced injury of vascular endotheliocytes. Our study is aimed at evaluating the effect of Andr on HG-induced HUVEC injury and the underlying mechanism. HUVECs were exposed to HG levels (33 mM) and treated with Andr (0, 12.5, 25, and 50 μM). Western blot analysis, real-time PCR, immunofluorescence staining, the scratch test, and the tube formation assay were performed to assess the effects of Andr. We discovered that Andr inhibited the inflammatory response (IL-1β, IL-6, and TNFα), decreased the apoptosis ratio and cell migration, and promoted tube formation in response to HG stimulation. Andr ameliorated the levels of phosphorylated PI3K (p-PI3K), phosphorylated AKT (p-AKT), and phosphorylated eNOS (p-eNOS). The expression of vascular endothelial growth factor (VEGF) protein, a vital factor in angiogenesis, was improved by Andr treatment under HG stimulation. LY294002 is a blocker of PI3K, MK-2206 2HCI (MK-2206) is a highly selective AKT inhibitor, and L-NAME is a suppressor of eNOS, all of which significantly reduce Andr-mediated protective effects in vitro. Hence, Andr may be involved in regulating HG-induced injury by activating PI3K/AKT-eNOS signalling in HUVECs.
Collapse
|
49
|
Pu Y, Wu D, Lu X, Yang L. Effects of GCN2/eIF2α on myocardial ischemia/hypoxia reperfusion and myocardial cells injury. Am J Transl Res 2019; 11:5586-5598. [PMID: 31632531 PMCID: PMC6789277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Myocardial ischemia/hypoxia-reperfusion injury is a common and severe cardiovascular disorder. General control non-derepressible 2 (GCN2) plays an important role in the role of cardiomyocyte glucose metabolism. Therefore, our study focused on the expression of GCN2/eIF2α in myocardial ischemia/hypoxia-reperfusion injury and its mechanism of myocardial cell injury. In the volunteers and patients with myocardial ischemia and hypoxia- reperfusion, the expression of GCN2 and eIF2α on serum were detected by RT-qPCR. The GCN2, eIF2α interference and GCN2 overexpression plasmids were constructed and transfected into cells. Then, the level of TNF-α, IL-1β, IL-6, IFN-γ were detected by ELISA and the level of ROS, MDA, LDH and SOD were measured by the corresponding kits, respectively. Besides, the expression of GCN2/eIF2 signaling pathway (p-eIF2α, ATF4, CHOP, UCP2 and eIF2α) and apoptosis-related proteins (Bax, Bcl-2 and cleaved caspase3) was detected by western blot. Flow cytometry was performed to value cell apoptosis. The expression of GCN2 is increased in oxygen-glucose deprivation/reoxygenation (OGD/R) model cells and GCN2 interference reduces the inflammation and oxidative stress in H9C2 cells after OGD/R. GCN2 interference reduced the level of apoptosis in OGD/R model cells and inhibited the expression of GCN2/eIF2α signaling pathway. We found that eIF2α interference could offset the effects of GCN2 overexpression on oxidative stress and apoptosis in H9C2 cells, and verified that GCN2 is produced by eIF2α phosphorylation. Together, GCN2/eIF2α signaling pathway plays an important role in myocardial ischemia/hypoxia-reperfusion injury, which could provide a new idea for the treatment of myocardial infarction on clinical.
Collapse
Affiliation(s)
- Yan Pu
- Department of Emergency Centre, Hanzhong Central HospitalHanzhong 723000, Shaanxi, China
| | - Dong Wu
- Department of Physical Emergency, Shaanxi Provincial People’s HospitalXi’an 710068, Shaanxi, China
| | - Xiaoe Lu
- Department of Critical Medicine, Shaanxi Provincial People’s HospitalXi’an 710068, Shaanxi, China
| | - Linjun Yang
- Department of Emergency, Hanzhong People’s HospitalHanzhong 723000, Shaanxi, China
| |
Collapse
|
50
|
Abstract
Heart failure and related morbidity and mortality are increasing at an alarming rate, in large part, because of increases in aging, obesity, and diabetes mellitus. The clinical outcomes associated with heart failure are considerably worse for patients with diabetes mellitus than for those without diabetes mellitus. In people with diabetes mellitus, the presence of myocardial dysfunction in the absence of overt clinical coronary artery disease, valvular disease, and other conventional cardiovascular risk factors, such as hypertension and dyslipidemia, has led to the descriptive terminology, diabetic cardiomyopathy. The prevalence of diabetic cardiomyopathy is increasing in parallel with the increase in diabetes mellitus. Diabetic cardiomyopathy is initially characterized by myocardial fibrosis, dysfunctional remodeling, and associated diastolic dysfunction, later by systolic dysfunction, and eventually by clinical heart failure. Impaired cardiac insulin metabolic signaling, mitochondrial dysfunction, increases in oxidative stress, reduced nitric oxide bioavailability, elevations in advanced glycation end products and collagen-based cardiomyocyte and extracellular matrix stiffness, impaired mitochondrial and cardiomyocyte calcium handling, inflammation, renin-angiotensin-aldosterone system activation, cardiac autonomic neuropathy, endoplasmic reticulum stress, microvascular dysfunction, and a myriad of cardiac metabolic abnormalities have all been implicated in the development and progression of diabetic cardiomyopathy. Molecular mechanisms linked to the underlying pathophysiological changes include abnormalities in AMP-activated protein kinase, peroxisome proliferator-activated receptors, O-linked N-acetylglucosamine, protein kinase C, microRNA, and exosome pathways. The aim of this review is to provide a contemporary view of these instigators of diabetic cardiomyopathy, as well as mechanistically based strategies for the prevention and treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Guanghong Jia
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.)
| | - Michael A Hill
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.)
| | - James R Sowers
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.).
| |
Collapse
|