1
|
Zhen J, Liu S, Liu L, Zheng X, Zhao G, Liang J, Xu A, Li C, Ren L, Wu J, Cheung BMY. Circulating fibroblast growth factor 21 is associated with blood pressure in the Chinese population: a community-based study. Ann Med 2025; 57:2500689. [PMID: 40356318 DOI: 10.1080/07853890.2025.2500689] [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: 09/23/2023] [Revised: 11/19/2024] [Accepted: 12/27/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Our research team previously found that fibroblast growth factor (FGF) 21, a circulating hormone, was significantly associated with atherosclerosis in human and animal models. The relationship between FGF21 and blood pressure (BP) is rarely studied in the Asian population. Therefore, we aimed to explore the relationship of FGF21 with BP in a Chinese population. METHODS We analysed data on 1051 participants from the Shenzhen-Hong Kong United Network on Cardiovascular Disease (SHUN-CVD) study. RESULTS The medians of FGF21 level were 355.1 pg/mL (IQR 234.3-574.8 pg/mL) for hypertensive patients and 253.5 (IQR 136.9-403.3 pg/mL) for non-hypertensive participants. Ln-transformed FGF21 level was associated with both systolic and diastolic BP (systolic BP: B = 4.45 [95% CI 3.41-5.49]; p < .001; diastolic BP: B = 2.72 [95% CI 2.03-3.42]; p < .001). After adjusting for sex, age, body mass index, hypercholesterolaemia, diabetes, alcohol consumption, smoking and physical activity, the association remained significant (systolic BP: B = 1.99 [95% CI 1.01-2.97]; p < .001; diastolic BP: B = 1.36 [95% CI 0.69-2.04]; p < .001). Serum FGF21 level was associated with hypertension (quartile 4 vs. quartile 1, OR = 4.19 [95% CI 2.65-6.61]; p for trend < .001). CONCLUSIONS This is the first study to elucidate the relationship of FGF21 with BP in the Asian population. FGF21 is significantly associated with BP. Besides its use as a biomarker, FGF21 may be a new drug target for hypertension treatment.
Collapse
Affiliation(s)
- Juanying Zhen
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shuyun Liu
- Department of Neurology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Lin Liu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Xiaodan Zheng
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guoru Zhao
- CAS Key Laboratory of Human-Machine Intelligence-Synergy Systems, Research Center for Neural Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianguo Liang
- 'Precision Health Research Center Company Limited, Hong Kong SAR, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chao Li
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Lijie Ren
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bernard Man Yung Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
2
|
Jalandhra GK, Srethbhakdi L, Davies J, Nguyen CC, Phan PT, Och Z, Ashok A, Lim KS, Phan HP, Do TN, Lovell NH, Rnjak-Kovacina J. Materials Advances in Devices for Heart Disease Interventions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2420114. [PMID: 40244561 DOI: 10.1002/adma.202420114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Heart disease encompasses a range of conditions that affect the heart, including coronary artery disease, arrhythmias, congenital heart defects, heart valve disease, and conditions that affect the heart muscle. Intervention strategies can be categorized according to when they are administered and include: 1) Monitoring cardiac function using sensor technology to inform diagnosis and treatment, 2) Managing symptoms by restoring cardiac output, electrophysiology, and hemodynamics, and often serving as bridge-to-recovery or bridge-to-transplantation strategies, and 3) Repairing damaged tissue, including myocardium and heart valves, when management strategies are insufficient. Each intervention approach and technology require specific material properties to function optimally, relying on materials that support their action and interface with the body, with new technologies increasingly depending on advances in materials science and engineering. This review explores material properties and requirements driving innovation in advanced intervention strategies for heart disease and highlights key examples of recent progress in the field driven by advances in materials research.
Collapse
Affiliation(s)
- Gagan K Jalandhra
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Lauryn Srethbhakdi
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - James Davies
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Chi Cong Nguyen
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Phuoc Thien Phan
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zachary Och
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Aditya Ashok
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Khoon S Lim
- School of Medical Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Hoang-Phuong Phan
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thanh Nho Do
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Nigel H Lovell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Tyree Institute of Health Engineering (IHealthE), University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Tyree Institute of Health Engineering (IHealthE), University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
3
|
Chen Y, Yang H, Wang Z. A dynamic study on serum biomarkers and kidney injury in a gosling model of hyperuricemia. Poult Sci 2025; 104:105144. [PMID: 40209470 PMCID: PMC12008651 DOI: 10.1016/j.psj.2025.105144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/30/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025] Open
Abstract
Hyperuricemia, a metabolic disorder characterized by elevated serum uric acid levels, poses significant challenges to poultry health and productivity. This study aimed to establish a hyperuricemia model in goslings through a multi-factorial induction approach using a high-purine, high-protein, and high-calcium diet, to investigate its dynamic progression, serological markers, and renal pathology. A total of 160 male Sanhua goslings were randomly assigned to control and model groups and fed either a basal diet or a high-purine, high-protein, and high-calcium diet for 18 days. Key findings revealed significant elevations in serum uric acid, creatinine, and urea nitrogen levels in the model group from day 5 onward, peaking on day 13 (p < 0.01). Activities of xanthine oxidase and adenosine deaminase were consistently higher in the model group, indicating enhanced purine metabolism and oxidative stress. Histopathological analysis showed progressive renal damage, including tubular degeneration, interstitial fibrosis, and glomerular injury, becoming evident by day 7 and severe by day 13. Ultrastructural examination further revealed mitochondrial dysfunction, podocyte effacement, and basement membrane disruption in the model group. Behavioral abnormalities, such as reduced mobility and the "shrinking neck" phenomenon, were observed from day 9, reflecting systemic disease severity. The dynamic expression of fibrosis-related markers, including Col-1, Col-3, and α-SMA, demonstrated the progression of renal fibrosis and extracellular matrix remodeling. These findings identify biomarkers for hyperuricemia progression and suggest potential therapeutic targets. This study establishes a comprehensive framework for understanding hyperuricemia-associated renal pathology and lays the groundwork for developing strategies to mitigate its impact on poultry health and productivity.
Collapse
Affiliation(s)
- Yuanjing Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Haiming Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Zhiyue Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China.
| |
Collapse
|
4
|
Chen L, Gao M, Ong SB, Gong G. Functions of FGF21 and its role in cardiac hypertrophy. J Adv Res 2025:S2090-1232(25)00148-1. [PMID: 40089060 DOI: 10.1016/j.jare.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND FGF21 is a stress-inducible hormone that operates in the autocrine or paracrine manner. Recent reports have revealed that FGF21 is highly expressed in cardiac hypertrophy to protect against heart injury and dysfunction. FGF21 is used to treat cardiac hypertrophy in mouse models. However, preclinical and clinical trials are restricted. AIM OF REVIEW This review mainly elucidates the diverse functions of FGF21 and explores the relationship between these functions and cardiac hypertrophy. It also discusses challenges and future perspectives in treating cardiac hypertrophy with FGF21. KEY SCIENTIFIC CONCEPTS OF REVIEW This review first illustrates the functions of FGF21, including energy metabolism, inflammation, oxidative stress, apoptosis, and autophagy. We also summarize vital functions and the underlying mechanisms through which FGF21 regulates the initiation and development of cardiac hypertrophy, connecting energy metabolism, inflammation, oxidative stress, apoptosis, and autophagy. Finally, we propose that FGF21 may be a potential therapeutic strategy for cardiac hypertrophy.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Meng Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), China
| | - Guohua Gong
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
5
|
Zhai C, Zhao Y, Zhang Z, Wang X, Li L, Li J. Mechanism of multifunctional adaptor protein SHARPIN regulating myocardial fibrosis and how SNP mutation affect the prognosis of myocardial infarction. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167467. [PMID: 39159699 DOI: 10.1016/j.bbadis.2024.167467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024]
Abstract
Myocardial fibrosis (MF) is characterized by the excessive deposition of extracellular matrix within the heart, often following a cardiovascular insult. SHARPIN, a protein implicated in fibrosis, has emerged as a potential therapeutic target. This study aimed to elucidate the molecular mechanisms of SHARPIN in MF and to investigate the influence of its single nucleotide polymorphism (SNP), rs117299156, on myocardial infarction (MI) patients. A mouse model of Angiotensin II (AngII)-induced MF was established in SHARPIN heterozygous (SHARPIN+/-) and wild-type mice. Adult mouse cardiac fibroblasts (CFs) were isolated and subjected to adenovirus-encapsulated SHARPIN short hairpin RNA (shRNA) infection. Transcriptomic analysis was performed on CFs from SHARPIN+/- and wild-type (WT) mice, complemented by single-cell sequencing data from human cardiac tissues. Additionally, the association between the rs117299156 mutation and cardiovascular events in MI patients was assessed. Our findings indicate that SHARPIN is predominantly expressed in CFs and is upregulated in fibrotic myocardium. Partial knockdown of SHARPIN in murine hearts mitigated AngII-induced cardiac dysfunction and MF. Furthermore, reduced SHARPIN expression in CFs attenuated TGF-β1-induced collagen synthesis, cell proliferation, and myofibroblast transformation. Notably, MI patients carrying the rs117299156_C allele exhibited a reduced incidence of stroke events compared to those without the mutation.
Collapse
Affiliation(s)
- Chao Zhai
- Division of Cardiology, Peking University First Hospital, Beijing 100034, China; Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Yixue Zhao
- Division of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Zhaoyu Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Xiaorui Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Li Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China.
| | - Jianping Li
- Division of Cardiology, Peking University First Hospital, Beijing 100034, China; Institute of Cardiovascular Disease, Peking University First Hospital, Beijing 100034, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing 100191, China.
| |
Collapse
|
6
|
Xu Z, Li H, Cao G, Li P, Zhou H, Sun Y. The protective role of brown adipose tissue in cardiac cell damage after myocardial infarction and heart failure. Lipids Health Dis 2024; 23:338. [PMID: 39415186 PMCID: PMC11481725 DOI: 10.1186/s12944-024-02326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024] Open
Abstract
Acute myocardial infarction (AMI) and related cardiovascular disease complications are the leading causes of mortality worldwide. Brown adipose tissue (BAT) is thermogenic and characterized by the uncoupling protein expression. Recent studies have found that in cardiovascular diseases, activated BAT can effectively improve the prognosis of AMI and concurrent heart failure through intercellular communication. However, a clear and systematic understanding of the myocardial protective mechanism of BAT after AMI is lacking, especially in the endocrine function of BAT. This review describes the effects of BAT on various cells in the heart after AMI. BAT plays a protective role on cardiac cells and fibroblasts during ischemia/reperfusion (I/R), myocardial remodeling, and myocardial fibrosis. This review also discusses the changes caused by BAT activation in different stages of heart failure. Finally, this review summarizes the treatment methods that target BAT to improve AMI. Further in-depth researches are still needed to clarify the underlying mechanism of the connection between BAT and different cells in cardiac tissue in order to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hong Li
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guojie Cao
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Panpan Li
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haitao Zhou
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Sun
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Zhang T, Jiang D, Zhang X, Chen L, Jiang J, Zhang C, Li S, Li Q. The role of nonmyocardial cells in the development of diabetic cardiomyopathy and the protective effects of FGF21: a current understanding. Cell Commun Signal 2024; 22:446. [PMID: 39327594 PMCID: PMC11426003 DOI: 10.1186/s12964-024-01842-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) represents a unique myocardial disease originating from diabetic metabolic disturbances that is characterized by myocardial fibrosis and diastolic dysfunction. While recent research regarding the pathogenesis and treatment of DCM has focused primarily on myocardial cells, nonmyocardial cells-including fibroblasts, vascular smooth muscle cells (VSMCs), endothelial cells (ECs), and immune cells-also contribute significantly to the pathogenesis of DCM. Among various therapeutic targets, fibroblast growth factor 21 (FGF21) has been identified as a promising agent because of its cardioprotective effects that extend to nonmyocardial cells. In this review, we aim to elucidate the role of nonmyocardial cells in DCM and underscore the potential of FGF21 as a therapeutic strategy for these cells.
Collapse
Affiliation(s)
- Tianyi Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Donghui Jiang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiao Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Shengbiao Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Qiuhong Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
8
|
Layek B. A Comprehensive Review of Xanthan Gum-Based Oral Drug Delivery Systems. Int J Mol Sci 2024; 25:10143. [PMID: 39337626 PMCID: PMC11431853 DOI: 10.3390/ijms251810143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Xanthan gum (XG) is an exopolysaccharide synthesized by the aerobic fermentation of simple sugars using Xanthomonas bacteria. It comprises a cellulosic backbone with a trisaccharide side chain connected to alternative glucose residues in the main backbone through α (1→3) linkage. XG dissolves readily in cold and hot water to produce a viscous solution that behaves like a pseudoplastic fluid. It shows excellent resistance to enzymatic degradation and great stability throughout a broad temperature, pH, or salt concentration range. Additionally, XG is nontoxic, biocompatible, and biodegradable, making it a suitable carrier for drug delivery. Furthermore, the carboxylic functions of pyruvate and glucuronic acid offer a considerable opportunity for chemical modification to meet the desired criteria for a specific application. Therefore, XG or its derivatives in conjunction with other polymers have frequently been studied as matrices for tablets, nanoparticles, microparticles, and hydrogels. This review primarily focuses on the applications of XG in various oral delivery systems over the past decade, including sustained-release formulations, gastroretentive dosage forms, and colon-targeted drug delivery. Source, production methods, and physicochemical properties relevant to drug delivery applications of XG have also been discussed.
Collapse
Affiliation(s)
- Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
9
|
Ding Y, Su J, Shan B, Fu X, Zheng G, Wang J, Wu L, Wang F, Chai X, Sun H, Zhang J. Brown adipose tissue-derived FGF21 mediates the cardioprotection of dexmedetomidine in myocardial ischemia/reperfusion injury. Sci Rep 2024; 14:18292. [PMID: 39112671 PMCID: PMC11306229 DOI: 10.1038/s41598-024-69356-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024] Open
Abstract
Brown adipose tissue (BAT) plays a critical role in regulating cardiovascular homeostasis through the secretion of adipokines, such as fibroblast growth factor 21 (FGF21). Dexmedetomidine (DEX) is a selective α2-adrenergic receptor agonist with a protection against myocardial ischemia/reperfusion injury (MI/RI). It remains largely unknown whether or not BAT-derived FGF21 is involved in DEX-induced cardioprotection in the context of MI/RI. Herein, we demonstrated that DEX alleviated MI/RI and improved heart function through promoting the release of FGF21 from interscapular BAT (iBAT). Surgical iBAT depletion or supplementation with a FGF21 neutralizing antibody attenuated the beneficial effects of DEX. AMPK/PGC1α signaling-induced fibroblast growth factor 21 (FGF21) release in brown adipocytes is required for DEX-mediated cardioprotection since blockade of the AMPK/PGC1α axis weakened the salutary effects of DEX. Co-culture experiments showed that DEX-induced FGF21 from brown adipocytes increased the resistance of cardiomyocytes to hypoxia/reoxygenation (H/R) injury via modulating the Keap1/Nrf2 pathway. Our results provided robust evidence that the BAT-cardiomyocyte interaction is required for DEX cardioprotection, and revealed an endocrine role of BAT in DEX-mediating protection of hearts against MIRI.
Collapse
Affiliation(s)
- Yi Ding
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Jiabao Su
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Beiying Shan
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Xiao Fu
- Laboratory of Metabolic and Inflammatory Diseases, Wuxi School of Medicine, Jiangnan University, No.1800, Lihu Road, Wuxi, 214125, People's Republic of China
| | - Guanli Zheng
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Jiwen Wang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Lixue Wu
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Fangming Wang
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, People's Republic of China
| | - Xiaoying Chai
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, People's Republic of China
| | - Haijian Sun
- Laboratory of Metabolic and Inflammatory Diseases, Wuxi School of Medicine, Jiangnan University, No.1800, Lihu Road, Wuxi, 214125, People's Republic of China.
| | - Jiru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China.
| |
Collapse
|
10
|
Wójcik M, Zieba DA, Bochenek J, Krawczyńska A, Barszcz M, Gajewska A, Antushevich H, Herman AP. The Effect of Endotoxin-Induced Inflammation on the Activity of the Somatotropic Axis in Sheep. Int J Inflam 2024; 2024:1057299. [PMID: 39149693 PMCID: PMC11325012 DOI: 10.1155/2024/1057299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/31/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024] Open
Abstract
The hypothalamic-pituitary-somatotropic (HPS) axis controls many physiological and pathophysiological processes. The phenomenon of insensitivity to growth hormone resistance (GHres) was previously reported to be due to the development of inflammation. Therefore, the primary aim of the study was to determine the impact of inflammation caused by lipopolysaccharides (LPS) on the secretory activity of the HPS axis in sheep. The further goal was to determine the effect of inflammatory factors on individual components involved in intracellular signal transduction to GH via the GH receptor (GHR). The research was carried out on 24 seasonal sheep kept under a short-day photoperiod, randomly divided into two groups. Before the experiment, the sheep estrous cycles were synchronized. The results of the current study in a sheep model showed that inflammation impairs the activity of the somatotropic axis. On the one hand, LPS injection stimulated (p < 0.01) GH secretion, and on the other hand, it reduced the liver's sensitivity to this hormone by directly reducing (p < 0.01) GHR expression and activating the GHR inhibitory signal transduction mechanism. A symptom of such an inhibitory postreceptor signaling pathway may be due to an increase in SOCS3 expression (p < 0.01). The effect of various inhibition pathways is a significant reduction in the expression of the main transcription activator IGF1-STAT5B (p < 0.05). The action of GHres in the liver resulted in the inhibition of IGF1 secretion, which in the long term may have negative consequences for growth and development. Our study suggests that disruption of the GH cell signaling pathway may be one of the important elements of the pathophysiology of inflammation. It can suppress growth and hepatic metabolism to spare energy expenditure.
Collapse
Affiliation(s)
- Maciej Wójcik
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| | - Dorota Anna Zieba
- Department of Nutrition and Animal Biotechnology, and Fisheries Faculty of Animal Sciences University of Agriculture in Krakow, Krakow 31-120, Poland
| | - Joanna Bochenek
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| | - Agata Krawczyńska
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| | - Marcin Barszcz
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| | - Alina Gajewska
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| | - Hanna Antushevich
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| | - Andrzej Przemysław Herman
- The Kielanowski Institute of Animal Physiology and Nutrition Polish Academy of Sciences, Instytucka 3 Street, Jabłonna 05-110, Poland
| |
Collapse
|
11
|
Xu G, Pan T, Li S, Guo J, Zhang Y, Xu Q, Chen R, Ma Y, Li Y. Mapping Single-Cell Transcriptomes of Endometrium Reveals Potential Biomarkers in Endometrial Cancer. Immunotargets Ther 2024; 13:349-366. [PMID: 39050484 PMCID: PMC11268782 DOI: 10.2147/itt.s470994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
Background The heterogeneity and dynamic changes of endometrial cells have a significant impact on health as they determine the normal function of the endometrium during the menstrual cycle. Dysfunction of the endometrium can lead to the occurrence of various gynecological diseases. Therefore, deconvolution of immune microenvironment that drives transcriptional programs throughout the menstrual cycle is key to understand regulatory biology of endometrium. Methods Herein, we comprehensively analyzed single-cell transcriptome of 59,397 cells across ten human endometrium samples and revealed the dynamic cellular heterogeneity throughout the menstrual cycle. Results We identified two perivascular cell subtypes, four epithelial subtypes and four fibroblast cell types in endometrium. Moreover, we inferred the cell type-specific transcription factor (TF) activities and linked critical TFs to transcriptional output of diverse immune cell types, highlighting the importance of transcriptional regulation in endometrium. Dynamic interactions between various types of cells in endometrium contribute to a range of biological pathways regulating differentiation of secretory. Integration of the molecular biomarkers identified in endometrium and bulk transcriptome of 535 endometrial cancers (EC), we revealed five RNA-based molecular subtypes of EC with highly intratumoral heterogeneity and different clinical manifestations. Mechanism analysis uncovered clinically relevant pathways for pathogenesis of EC. Conclusion In summary, our results revealed the dynamic immune microenvironment of endometrium and provided novel insights into future development of RNA-based treatments for endometriosis and endometrial carcinoma.
Collapse
Affiliation(s)
- Gang Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China, Harbin, 150081, People’s Republic of China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Tao Pan
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Si Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Jing Guo
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Ya Zhang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Qi Xu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Renwei Chen
- Hainan Women and Children’s Medical Center, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
| | - Yongsheng Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, National Center for International Research, The First Affiliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
12
|
Qi J, Wu Y, Guo Z, Zhu S, Xiong J, Hu F, Liang X, Ye X. Fibroblast growth factor 21 alleviates idiopathic pulmonary fibrosis by inhibiting PI3K-AKT-mTOR signaling and stimulating autophagy. Int J Biol Macromol 2024; 273:132896. [PMID: 38851619 DOI: 10.1016/j.ijbiomac.2024.132896] [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: 05/12/2023] [Revised: 05/20/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive pulmonary disease with an unclear pathogenesis and no available specific drug treatment. The principal etiological factors are lung inflammation caused by environmental factors, damage to alveolar epithelial cells, leading to epithelial-mesenchymal transition (EMT), and the abnormal proliferation of fibroblasts. Here, we have demonstrated that fibroblast growth factor 21 (FGF21) ameliorates IPF via the autophagy pathway. We administered FGF21 to bleomycin (BLM)-treated mice, which ameliorated their defects in lung function, reduced the accumulation of collagen, restored tissue structure, reduced the deposition of hydroxyproline, reduced the expression of collagen I and α-SMA and increased the expression of E-cadherin. The expression of LC3BII and the number of autophagosomes were significantly higher in the lungs. The expression of AKT and mTOR was significantly reduced by FGF21 treatment. We also determined the effects of FGF21 in A549 cells treated with TGF-β, and found that FGF21 significantly inhibits activation of the AKT signaling pathway, thereby reducing TGF-β-induced EMT and preventing the uncontrolled proliferation of fibroblasts. We conclude that FGF21 ameliorates IPF by inhibiting the PI3K-AKT-mTOR signaling pathway and activating autophagy, which provides a theoretical basis for FGF21 to be used for the treatment of IPF.
Collapse
Affiliation(s)
- Jianying Qi
- School of chemical engineering, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Yuanyuan Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Zhimou Guo
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian 116023, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jingjing Xiong
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Fei Hu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian 116023, China.
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China.
| |
Collapse
|
13
|
Ghanem M, Archer G, Crestani B, Mailleux AA. The endocrine FGFs axis: A systemic anti-fibrotic response that could prevent pulmonary fibrogenesis? Pharmacol Ther 2024; 259:108669. [PMID: 38795981 DOI: 10.1016/j.pharmthera.2024.108669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal disease for which therapeutic options are limited, with an unmet need to identify new therapeutic targets. IPF is thought to be the consequence of repeated microlesions of the alveolar epithelium, leading to aberrant epithelial-mesenchymal communication and the accumulation of extracellular matrix proteins. The reactivation of developmental pathways, such as Fibroblast Growth Factors (FGFs), is a well-described mechanism during lung fibrogenesis. Secreted FGFs with local paracrine effects can either exert an anti-fibrotic or a pro-fibrotic action during this pathological process through their FGF receptors (FGFRs) and heparan sulfate residues as co-receptors. Among FGFs, endocrine FGFs (FGF29, FGF21, and FGF23) play a central role in the control of metabolism and tissue homeostasis. They are characterized by a low affinity for heparan sulfate, present in the cell vicinity, allowing them to have endocrine activity. Nevertheless, their interaction with FGFRs requires the presence of mandatory co-receptors, alpha and beta Klotho proteins (KLA and KLB). Endocrine FGFs are of growing interest for their anti-fibrotic action during liver, kidney, or myocardial fibrosis. Innovative therapies based on FGF19 or FGF21 analogs are currently being studied in humans during liver fibrosis. Recent data report a similar anti-fibrotic action of endocrine FGFs in the lung, suggesting a systemic regulation of the pulmonary fibrotic process. In this review, we summarize the current knowledge on the protective effect of endocrine FGFs during the fibrotic processes, with a focus on pulmonary fibrosis.
Collapse
Affiliation(s)
- Mada Ghanem
- Université Paris Cité, Inserm, Physiopathologie et Épidémiologie des Maladies Respiratoires, F-75018 Paris, France
| | - Gabrielle Archer
- Université Paris Cité, Inserm, Physiopathologie et Épidémiologie des Maladies Respiratoires, F-75018 Paris, France
| | - Bruno Crestani
- Université Paris Cité, Inserm, Physiopathologie et Épidémiologie des Maladies Respiratoires, F-75018 Paris, France; Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, FHU APOLLO, Paris, France
| | - Arnaud A Mailleux
- Université Paris Cité, Inserm, Physiopathologie et Épidémiologie des Maladies Respiratoires, F-75018 Paris, France.
| |
Collapse
|
14
|
Li R, Liu X. FGF21 Inhibits Hypoxia/Reoxygenation-induced Renal Tubular Epithelial Cell Injury by Regulating the PPARγ/NF-κB Signaling Pathway. Cell Biochem Biophys 2024; 82:909-918. [PMID: 38459267 DOI: 10.1007/s12013-024-01242-8] [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] [Accepted: 02/23/2024] [Indexed: 03/10/2024]
Abstract
As a predominant trigger of acute kidney injury, renal ischemia-reperfusion injury can cause permanent renal impairment, and the effective therapies are lacking. Fibroblast growth factor 21 (FGF21) plays a critical regulatory role in a variety of biological activities. This study was conducted to explore the functional of FGF21 in renal ischemia-reperfusion injury and to discuss the hidden reaction mechanism. To simulate renal ischemia-reperfusion injury in vitro, HK2 cells were induced by hypoxia/reoxygenation (H/R). The effects of FGF21 on H/R-induced HK2 cell viability were evaluated utilizing cell counting kit-8 (CCK-8). The levels of lactate dehydrogenase (LDH) and inflammatory cytokines in H/R-induced HK2 cells were assessed by means of LDH assay and enzyme-linked immunosorbent assay (ELISA). The levels of oxidative stress markers were appraised with corresponding assay kits and western blot was applied to estimate the expressions of oxidative stress-related proteins. The apoptosis of H/R-induced HK2 cells was assessed by virtue of flow cytometry. The expressions of apoptosis- and PPARγ/NF-κB signaling pathway-related proteins were evaluated with western blot. To discuss the reaction mechanism of PPARγ/NF-κB pathway in H/R-induced HK2 cells, PPARγ inhibitor GW9662 was employed to treat cells and the above experiments were then conducted again. This study found that FGF21 treatment inhibited the inflammatory response, oxidative stress and apoptosis in H/R-induced HK2 cells. Moreover, FGF21 regulated PPARγ/NF-κB signaling pathway and GW9662 partially reversed the impacts of FGF21 on the inflammatory response, oxidative stress and apoptosis in H/R-exposed HK2 cells. Collectively, FGF21 protected against H/R-induced renal tubular epithelial cell injury by regulating the PPARγ/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ruixue Li
- Nephrology Department, The People's Hospital of Yubei District of Chongqing, Chongqing, 401120, PR China.
| | - Xi Liu
- Nephrology Department, The People's Hospital of Yubei District of Chongqing, Chongqing, 401120, PR China
| |
Collapse
|
15
|
Berezin OO, Berezina TA, Hoppe UC, Lichtenauer M, Berezin AE. Diagnostic and predictive abilities of myokines in patients with heart failure. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:45-98. [PMID: 39059994 DOI: 10.1016/bs.apcsb.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Myokines are defined as a heterogenic group of numerous cytokines, peptides and metabolic derivates, which are expressed, synthesized, produced, and released by skeletal myocytes and myocardial cells and exert either auto- and paracrine, or endocrine effects. Previous studies revealed that myokines play a pivotal role in mutual communications between skeletal muscles, myocardium and remote organs, such as brain, vasculature, bone, liver, pancreas, white adipose tissue, gut, and skin. Despite several myokines exert complete divorced biological effects mainly in regulation of skeletal muscle hypertrophy, residential cells differentiation, neovascularization/angiogenesis, vascular integrity, endothelial function, inflammation and apoptosis/necrosis, attenuating ischemia/hypoxia and tissue protection, tumor growth and malignance, for other occasions, their predominant effects affect energy homeostasis, glucose and lipid metabolism, adiposity, muscle training adaptation and food behavior. Last decade had been identified 250 more myokines, which have been investigating for many years further as either biomarkers or targets for heart failure management. However, only few myokines have been allocated to a promising tool for monitoring adverse cardiac remodeling, ischemia/hypoxia-related target-organ dysfunction, microvascular inflammation, sarcopenia/myopathy and prediction for poor clinical outcomes among patients with HF. This we concentrate on some most plausible myokines, such as myostatin, myonectin, brain-derived neurotrophic factor, muslin, fibroblast growth factor 21, irisin, leukemia inhibitory factor, developmental endothelial locus-1, interleukin-6, nerve growth factor and insulin-like growth factor-1, which are suggested to be useful biomarkers for HF development and progression.
Collapse
Affiliation(s)
- Oleksandr O Berezin
- Luzerner Psychiatrie AG, Department of Senior Psychiatrie, St. Urban, Switzerland
| | - Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, Ukraine
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
16
|
Zhang Y, Song Q, E S, Guan X, Zhang Z, Juan Z, Sun X, Liang Y. RNA-seq based transcriptomic map reveals multiple pathways of necroptosis in treating myocardial ischemia reperfusion injury. Gene 2024; 906:148217. [PMID: 38341002 DOI: 10.1016/j.gene.2024.148217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/19/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024]
Abstract
Necroptosis has been shown to contribute to myocardial ischemia reperfusion injury (MIRI). This study aims to gain new insights into the signaling pathway of necroptosis in rat MIRI using RNA sequencing. MIRI was induced in male rats by ligating the left anterior descending coronary artery for 30 min, followed by reperfusion for 120 min. RNA sequencing was performed to obtain mRNA profiles of MIRI group and MIRI group treated with necrostatin-1 (Nec-1,an inhibitor of necroptosis). Differentially expressed genes (DEGs) were then identified. The DEGs were prominently enriched in the TNF-α signaling pathway, the MAPK signaling pathway and cytokine-cytokine receptor pathways. The majority of the results were associated with genes like Thumpd3,Egr2,Dot1l,Cyp1a1,Dbnl,which primarily regulate inflammatory response and apoptosis, particularly in myocardium. The above results suggested that Nec-1 might be involved in the regulation of necroptosis and the inflammatory response through the above-mentioned genes.
Collapse
Affiliation(s)
- Yijia Zhang
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Qingbiao Song
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Sihan E
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Xuehao Guan
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Zhiyu Zhang
- Department of Orthopedics, Affiliated Hospital of Weifang Medical University, Weifang 261035, China
| | - Zhaodong Juan
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Xiaotong Sun
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Yingxia Liang
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
17
|
Jia Y, Zhang RN, Li YJ, Guo BY, Wang JL, Liu SY. Bioinformatics analysis and identification of potential key genes and pathways in the pathogenesis of nonischemic cardiomyopathy. Medicine (Baltimore) 2024; 103:e37898. [PMID: 38669428 PMCID: PMC11049792 DOI: 10.1097/md.0000000000037898] [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: 02/09/2023] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Nonischemic cardiomyopathy (NICM) is a major cause of advanced heart failure, and the morbidity and mortality associated with NICM are serious medical problems. However, the etiology of NICM is complex and the related mechanisms involved in its pathogenesis remain unclear. The microarray datasets GSE1869 and GSE9128 retrieved from the Gene Expression Omnibus database were used to identify differentially expressed genes (DEGs) between NICM and normal samples. The co-expressed genes were identified using Venn diagrams. Kyoto Encyclopedia of Genes and Genomes pathway analyses and gene ontology enrichment were used to clarify biological functions and signaling pathways. Analysis of protein-protein interaction networks using Search Tool for the Retrieval of Interacting Genes/Proteins online to define the hub genes associated with NICM pathogenesis. A total of 297 DEGs were identified from GSE1869, 261 of which were upregulated genes and 36 were downregulated genes. A total of 360 DEGs were identified from GSE9128, 243 of which were upregulated genes and 117 were downregulated genes. In the 2 datasets, the screening identified 36 co-expressed DEGs. Kyoto Encyclopedia of Genes and Genomes pathway and gene ontology analysis showed that DEGs were mainly enriched in pantothenate and CoA biosynthesis, beta-alanine metabolism, kinetochore, G-protein beta/gamma-subunit complex, and other related pathways. The PPI network analysis revealed that DUSP6, EGR1, ZEB2, and XPO1 are the 4 hub genes of interest in the 2 datasets. Bioinformatics analysis of hub genes and key signaling pathways is an effective way to elucidate the mechanisms involved in the development of NICM. The results will facilitate further studies on the pathogenesis and therapeutic targets of NICM.
Collapse
Affiliation(s)
- Yan Jia
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Rui-Ning Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong-Jun Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bing-Yan Guo
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jian-Long Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Su-Yun Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
18
|
Tian G, Li J, Wang W, Zhou L. FGF12 restrains mitochondria-dependent ferroptosis in doxorubicin-induced cardiomyocytes through the activation of FGFR1/AMPK/NRF2 signaling. Drug Dev Res 2024; 85:e22149. [PMID: 38349269 DOI: 10.1002/ddr.22149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 02/15/2024]
Abstract
Fibroblast growth factor-12 (FGF12) has been reported to play important role in regulating heart diseases. We aimed to explore the role of FGF12 in doxorubicin (DOX)-induced myocardial injury. DOX-induced mice and DOX-induced HL-1 cells were used as the myocardial injury in vivo and in vitro. Then, FGF12, Anp, Bnp, and Myh7 expression was detected. The pathological injury in myocardium tissue was observed by H&E staining. The levels of markers related to myocardial damage and oxidative stress were assessed. Then, immunohistochemistry and immunofluorescence staining were used to detect FGF12 and 4-HNE expression. Ferroptosis were detected by Prussian blue staining and western blot. The FGFR1/AMPK/NRF2 signaling was measured by western blot. FGF12 expression was downregulated in DOX-induced mice myocardium tissues. FGF12 overexpression alleviated DOX-induced myocardial tissue pathological injury and reduced Anp, Bnp, and Myh7 expression. Additionally, the levels of CK-MB, LDH and cTnT in serum were decreased after FGF12 upregulation in DOX-induced mice. Moreover, FGF12 overexpression reduced the levels of ROS, MDA, and 4-HNE but increased SOD and GSH-Px activities. Meanwhile, FGF12 led to less deposition of iron ion, decreased ACSL4, PTGS2 and increased GPX4, FTH1 expression. Additionally, FGF12 activated the expressions of FGFR1, p-AMPK, and NRF2. Moreover, FGFR1 silencing reversed the protective effects of FGF12 overexpression on cell viability, oxidative stress, ferroptosis, and FGFR1/AMPK/NRF2 pathway. To sum up, FGF12 inhibited mitochondria-dependent ferroptosis in cardiomyocytes induced by DOX through activation of FGFR1/AMPK/NRF2 signaling. These findings clarify a new mechanism of DOX-induced cardiac injury and provide a promising target to limit the disease development.
Collapse
Affiliation(s)
- Ge Tian
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jing Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wenjie Wang
- Department of General Practice, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lina Zhou
- Department of Geriatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
19
|
Wu L, Yin W, Wen J, Wang S, Li H, Wang X, Zhang W, Duan S, Zhu Q, Gao E, Wu S, Zhan B, Zhou R, Yang X. Excretory/secretory products from Trichinella spiralis adult worms ameliorate myocardial infarction by inducing M2 macrophage polarization in a mouse model. Parasit Vectors 2023; 16:362. [PMID: 37845695 PMCID: PMC10577921 DOI: 10.1186/s13071-023-05930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Ischemia-induced inflammatory response is the main pathological mechanism of myocardial infarction (MI)-caused heart tissue injury. It has been known that helminths and worm-derived proteins are capable of modulating host immune response to suppress excessive inflammation as a survival strategy. Excretory/secretory products from Trichinella spiralis adult worms (Ts-AES) have been shown to ameliorate inflammation-related diseases. In this study, Ts-AES were used to treat mice with MI to determine its therapeutic effect on reducing MI-induced heart inflammation and the immunological mechanism involved in the treatment. METHODS The MI model was established by the ligation of the left anterior descending coronary artery, followed by the treatment of Ts-AES by intraperitoneal injection. The therapeutic effect of Ts-AES on MI was evaluated by measuring the heart/body weight ratio, cardiac systolic and diastolic functions, histopathological change in affected heart tissue and observing the 28-day survival rate. The effect of Ts-AES on mouse macrophage polarization was determined by stimulating mouse bone marrow macrophages in vitro with Ts-AES, and the macrophage phenotype was determined by flow cytometry. The protective effect of Ts-AES-regulated macrophage polarization on hypoxic cardiomyocytes was determined by in vitro co-culturing Ts-AES-induced mouse bone marrow macrophages with hypoxic cardiomyocytes and cardiomyocyte apoptosis determined by flow cytometry. RESULTS We observed that treatment with Ts-AES significantly improved cardiac function and ventricular remodeling, reduced pathological damage and mortality in mice with MI, associated with decreased pro-inflammatory cytokine levels, increased regulatory cytokine expression and promoted macrophage polarization from M1 to M2 type in MI mice. Ts-AES-induced M2 macrophage polarization also reduced apoptosis of hypoxic cardiomyocytes in vitro. CONCLUSIONS Our results demonstrate that Ts-AES ameliorates MI in mice by promoting the polarization of macrophages toward the M2 type. Ts-AES is a potential pharmaceutical agent for the treatment of MI and other inflammation-related diseases.
Collapse
Affiliation(s)
- Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Wenhui Yin
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Jutai Wen
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Shuying Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Weixiao Zhang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Shuyao Duan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Qiuyu Zhu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Erhe Gao
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Shili Wu
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
20
|
Cao MJ, Shi FH, Yu BB, Ma XC, Zhang C, Xu L, Jiang YH, Ge H, Shen L, Pu J. Influence and mechanism of sodium-glucose cotransporter-2 inhibitors on the cardiac function: study protocol for a prospective cohort study. Front Endocrinol (Lausanne) 2023; 14:1199960. [PMID: 37538793 PMCID: PMC10395085 DOI: 10.3389/fendo.2023.1199960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/29/2023] [Indexed: 08/05/2023] Open
Abstract
Background Acute myocardial infarction (AMI) poses a significant threat to cardiovascular diseases (CVDs), leading to a high risk of heart failure (HF) and cardiovascular death. Growing evidence has unveiled the potential of sodium-glucose cotransporter-2 (SGLT2) inhibitors to improve cardiovascular outcomes in patients with CVD regardless of diabetes, but there is limited evidence in AMI patients. Furthermore, it is controversial whether the effects can be ascribed to the amelioration of left ventricular (LV) function, which further complicates the understanding of their underlying mechanism. Methods This study is a prospective, phase IV, open-label, parallel group, single-center trial conducted in a large tertiary teaching hospital in China. A total of 120 patients with AMI and type 2 diabetes mellitus (T2DM) will be included. Those who received SGLT2 inhibitors are considered as the experimental group, and those taking other antidiabetic agents are considered as the control group. The primary outcome is change in LV end-systolic volume index (LVESVi) measured by cardiac magnetic resonance (CMR) imaging from baseline during 1-year follow-up period. Secondary outcomes include other LV parameters such as LV mass, LV volume, and LV ejection fraction (EF); quality of life and functional capacity such as Kansas City Cardiomyopathy Questionnaire overall summary score (KCCQ-OS) and EuroQol-5 dimension (EQ-5D); biomarkers associated with diagnostic parameters of AMI and possible mechanisms on cardiovascular protection, such as creatine kinase, troponin T (TnT) level, troponin I (TnI) level, soluble suppression of tumorigenicity-2 (sST2), galectin-3 (Gal-3), fibroblast growth factor 21 (FGF21), and microRNA (miRNA) level. Discussion This study aims to investigate whether SGLT2 inhibitors could improve LV function by measuring CMR, quality of life, and functional capacity in patients with AMI in real-world settings, providing evidence on the underlying mechanism of SGLT2 inhibitors on cardioprotection. Clinical trial registration https://www.chictr.org.cn/showproj.html?proj=173672, identifier ChiCTR2200065792.
Collapse
Affiliation(s)
- Min-Jia Cao
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang-Hong Shi
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin-Bin Yu
- Department of Pharmacy, Huangyan Hospital of Wenzhou Medical University, Taizhou First People’s Hospital, Zhejiang, China
| | - Xue-Chen Ma
- Department of Pharmacy, The Third People’s Hospital of Xining, Xining, China
| | - Chen Zhang
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Xu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Hong Jiang
- Department of Endocrinology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Heng Ge
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Shen
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Zhao Z, Cui X, Liao Z. Mechanism of fibroblast growth factor 21 in cardiac remodeling. Front Cardiovasc Med 2023; 10:1202730. [PMID: 37416922 PMCID: PMC10322220 DOI: 10.3389/fcvm.2023.1202730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Cardiac remodeling is a basic pathological process that enables the progression of multiple cardiac diseases to heart failure. Fibroblast growth factor 21 is considered a regulator in maintaining energy homeostasis and shows a positive role in preventing damage caused by cardiac diseases. This review mainly summarizes the effects and related mechanisms of fibroblast growth factor 21 on pathological processes associated with cardiac remodeling, based on a variety of cells of myocardial tissue. The possibility of Fibroblast growth factor 21 as a promising treatment for the cardiac remodeling process will also be discussed.
Collapse
Affiliation(s)
- Zeyu Zhao
- Queen Mary College, Nanchang University, Nanchang, China
| | - Xuemei Cui
- Fourth Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhangping Liao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology School of Pharmaceutical Science, Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Gil-Cabrerizo P, Scaccheti I, Garbayo E, Blanco-Prieto MJ. Cardiac tissue engineering for myocardial infarction treatment. Eur J Pharm Sci 2023; 185:106439. [PMID: 37003408 DOI: 10.1016/j.ejps.2023.106439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Myocardial infarction is one of the major causes of morbidity and mortality worldwide. Current treatments can relieve the symptoms of myocardial ischemia but cannot repair the necrotic myocardial tissue. Novel therapeutic strategies based on cellular therapy, extracellular vesicles, non-coding RNAs and growth factors have been designed to restore cardiac function while inducing cardiomyocyte cycle re-entry, ensuring angiogenesis and cardioprotection, and preventing ventricular remodeling. However, they face low stability, cell engraftment issues or enzymatic degradation in vivo, and it is thus essential to combine them with biomaterial-based delivery systems. Microcarriers, nanocarriers, cardiac patches and injectable hydrogels have yielded promising results in preclinical studies, some of which are currently being tested in clinical trials. In this review, we cover the recent advances made in cellular and acellular therapies used for cardiac repair after MI. We present current trends in cardiac tissue engineering related to the use of microcarriers, nanocarriers, cardiac patches and injectable hydrogels as biomaterial-based delivery systems for biologics. Finally, we discuss some of the most crucial aspects that should be addressed in order to advance towards the clinical translation of cardiac tissue engineering approaches.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ilaria Scaccheti
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| |
Collapse
|
23
|
Yin X, Yin X, Pan X, Zhang J, Fan X, Li J, Zhai X, Jiang L, Hao P, Wang J, Chen Y. Post-myocardial infarction fibrosis: Pathophysiology, examination, and intervention. Front Pharmacol 2023; 14:1070973. [PMID: 37056987 PMCID: PMC10086160 DOI: 10.3389/fphar.2023.1070973] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Cardiac fibrosis plays an indispensable role in cardiac tissue homeostasis and repair after myocardial infarction (MI). The cardiac fibroblast-to-myofibroblast differentiation and extracellular matrix collagen deposition are the hallmarks of cardiac fibrosis, which are modulated by multiple signaling pathways and various types of cells in time-dependent manners. Our understanding of the development of cardiac fibrosis after MI has evolved in basic and clinical researches, and the regulation of fibrotic remodeling may facilitate novel diagnostic and therapeutic strategies, and finally improve outcomes. Here, we aim to elaborate pathophysiology, examination and intervention of cardiac fibrosis after MI.
Collapse
Affiliation(s)
- Xiaoying Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinxin Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Pan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyu Zhang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinhui Fan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiaxin Li
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoxuan Zhai
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lijun Jiang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Panpan Hao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiali Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
24
|
Yang L, Nao J. Focus on Alzheimer's Disease: The Role of Fibroblast Growth Factor 21 and Autophagy. Neuroscience 2023; 511:13-28. [PMID: 36372296 DOI: 10.1016/j.neuroscience.2022.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/24/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Alzheimer's disease (AD) is a disorder of the central nervous system that is typically marked by progressive cognitive impairment and memory loss. Amyloid β plaque deposition and neurofibrillary tangles with hyperphosphorylated tau are the two hallmark pathologies of AD. In mammalian cells, autophagy clears aberrant protein aggregates, thus maintaining proteostasis as well as neuronal health. Autophagy affects production and metabolism of amyloid β and accumulation of phosphorylated tau proteins, whose malfunction can lead to the progression of AD. On the other hand, defective autophagy has been found to induce the production of the neuroprotective factor fibroblast growth factor 21 (FGF21), although the underlying mechanism is unclear. In this review, we highlight the significance of aberrant autophagy in the pathogenesis of AD, discuss the possible mechanisms by which defective autophagy induces FGF21 production, and analyze the potential of FGF21 in the treatment of AD. The findings provide some insights into the potential role of FGF21 and autophagy in the pathogenesis of AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
25
|
Gu L, Jiang W, Jiang W, Xu Z, Li W, Zhang H. Elevated serum FGF21 levels predict heart failure during hospitalization of STEMI patients after emergency percutaneous coronary intervention. PeerJ 2023; 11:e14855. [PMID: 36778154 PMCID: PMC9910186 DOI: 10.7717/peerj.14855] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/14/2023] [Indexed: 02/08/2023] Open
Abstract
Background Fibroblast growth factor 21 (FGF21) has multiple cardioprotective effects including modulation of glucolipid metabolism, anti-inflammation, and anti-oxidative stress, but its association with the heart failure during hospitalization in patients with ST-segment elevation myocardial infarction (STEMI) undergoing emergency percutaneous coronary intervention (PCI) has not been reported. Methods A total of 348 STEMI patients treated with emergency PCI were included from January 2016 to December 2018. Relevant biochemical indicators were measured by central laboratory. Serum FGF21 levels were measured by ELISA. The occurrence of heart failure during hospitalization was recorded. Patients' cardiac function was assessed by echocardiography. Results Serum FGF21 levels were significantly higher in the STEMI group with heart failure than in the group without heart failure (249.95 ± 25.52 vs. 209.98 ± 36.35, P < 0.001). Serum FGF21 levels showed a strong positive correlation with N-terminal precursor B-type natriuretic peptide (NT-proBNP) in STEMI patients (r = 0.749, P < 0.001). FGF21 was found to be an independent risk factor for the development of heart failure during hospitalization in STEMI patients by binary logistic regression analysis. The area under curve (AUC) for FGF21 to predict the development of heart failure during hospitalization in STEMI patients was 0.816 (95% CI [0.770-0.863]) according to the receiver operating characteristic (ROC) curve analysis. Conclusion Elevated serum FGF21 levels have been shown to be a strong predictor of heart failure during hospitalization in patients with STEMI after emergency PCI.
Collapse
|
26
|
Arias-Calderón M, Casas M, Balanta-Melo J, Morales-Jiménez C, Hernández N, Llanos P, Jaimovich E, Buvinic S. Fibroblast growth factor 21 is expressed and secreted from skeletal muscle following electrical stimulation via extracellular ATP activation of the PI3K/Akt/mTOR signaling pathway. Front Endocrinol (Lausanne) 2023; 14:1059020. [PMID: 36909316 PMCID: PMC9997036 DOI: 10.3389/fendo.2023.1059020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a hormone involved in the regulation of lipid, glucose, and energy metabolism. Although it is released mainly from the liver, in recent years it has been shown that it is a "myokine", synthesized in skeletal muscles after exercise and stress conditions through an Akt-dependent pathway and secreted for mediating autocrine and endocrine roles. To date, the molecular mechanism for the pathophysiological regulation of FGF21 production in skeletal muscle is not totally understood. We have previously demonstrated that muscle membrane depolarization controls gene expression through extracellular ATP (eATP) signaling, by a mechanism defined as "Excitation-Transcription coupling". eATP signaling regulates the expression and secretion of interleukin 6, a well-defined myokine, and activates the Akt/mTOR signaling pathway. This work aimed to study the effect of electrical stimulation in the regulation of both production and secretion of skeletal muscle FGF21, through eATP signaling and PI3K/Akt pathway. Our results show that electrical stimulation increases both mRNA and protein (intracellular and secreted) levels of FGF21, dependent on an extracellular ATP signaling mechanism in skeletal muscle. Using pharmacological inhibitors, we demonstrated that FGF21 production and secretion from muscle requires the activation of the P2YR/PI3K/Akt/mTOR signaling pathway. These results confirm skeletal muscle as a source of FGF21 in physiological conditions and unveil a new molecular mechanism for regulating FGF21 production in this tissue. Our results will allow to identify new molecular targets to understand the regulation of FGF21 both in physiological and pathological conditions, such as exercise, aging, insulin resistance, and Duchenne muscular dystrophy, all characterized by an alteration in both FGF21 levels and ATP signaling components. These data reinforce that eATP signaling is a relevant mechanism for myokine expression in skeletal muscle.
Collapse
Affiliation(s)
- Manuel Arias-Calderón
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Mariana Casas
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
| | - Julián Balanta-Melo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- School of Dentistry, Faculty of Health, Universidad del Valle, Cali, Colombia
| | - Camilo Morales-Jiménez
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Department of Basic Sciences of Health, Faculty of Health Sciences, Pontificia Universidad Javeriana, Cali, Colombia
| | - Nadia Hernández
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
- *Correspondence: Sonja Buvinic,
| |
Collapse
|
27
|
Martins FF, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Brown adipose tissue as an endocrine organ: updates on the emerging role of batokines. Horm Mol Biol Clin Investig 2022:hmbci-2022-0044. [DOI: 10.1515/hmbci-2022-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/20/2022] [Indexed: 11/15/2022]
Abstract
Abstract
Brown adipose tissue (BAT) remains active in adults, oxidizing fatty acids or glucose and releasing energy in the form of heat. Brown adipocytes and enhanced thermogenesis are targets for treating obesity and its comorbidities. BAT shows high synthesis activity and secretes several signaling molecules. The brown adipokines, or batokines, take action in an autocrine, paracrine, and endocrine manner. Batokines have a role in the homeostasis of the cardiovascular system, central nervous system, white adipose tissue, liver, and skeletal muscle and exert beneficial effects on BAT. The systemic function of batokines gives BAT an endocrine organ profile. Besides, the batokines Fibroblast Growth Factor-21, Vascular Endothelial Growth Factor A, Bone Morphogenetic Protein 8, Neuregulin 4, Myostatin, and Interleukin-6 emerge as targets to treat obesity and its comorbidities, deserving attention. This review outlines the role of six emerging batokines on BAT and their cross-talk with other organs, focusing on their physiological significance and diet-induced changes.
Collapse
Affiliation(s)
- Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases , Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases , Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases , Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases , Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro , Rio de Janeiro , Brazil
| |
Collapse
|
28
|
Schmitz T, Harmel E, Heier M, Peters A, Linseisen J, Meisinger C. Inflammatory plasma proteins predict short-term mortality in patients with an acute myocardial infarction. J Transl Med 2022; 20:457. [PMID: 36209229 PMCID: PMC9547640 DOI: 10.1186/s12967-022-03644-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/18/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate the association between inflammatory markers and 28-day mortality in patients with ST-elevation myocardial infarction (STEMI). METHODS In 398 STEMI patients recorded between 2009 and 2013 by the population-based Myocardial Infarction Registry Augsburg, 92 protein biomarkers were measured in admission arterial blood samples using the OLINK inflammatory panel. In multivariable-adjusted logistic regression models, the association between each marker and 28-day mortality was investigated. The values of the biomarkers most significantly associated with mortality were standardized and summarized to obtain a prediction score for 28-day mortality. The predictive ability of this biomarker score was compared to the established GRACE score using ROC analysis. Finally, a combined total score was generated by adding the standardized biomarker score to the standardized GRACE score. RESULTS The markers IL-6, IL-8, IL-10, FGF-21, FGF-23, ST1A1, MCP-1, 4E-BP1, and CST5 were most significantly associated with 28-day mortality, each with FDR-adjusted (false discovery rate adjusted) p-values of < 0.01 in the multivariable logistic regression model. In a ROC analysis, the biomarker score and the GRACE score showed comparable predictive ability for 28-day mortality (biomarker score AUC: 0.7859 [CI: 0.6735-0.89], GRACE score AUC: 0.7961 [CI: 0.6965-0.8802]). By combining the biomarker score and the Grace score, the predictive ability improved with an AUC of 0.8305 [CI: 0.7269-0.9187]. A continuous Net Reclassification Improvement (cNRI) of 0.566 (CI: 0.192-0.94, p-value: 0.003) and an Integrated Discrimination Improvement (IDI) of 0.083 ((CI: 0.016-0.149, p-value: 0.015) confirmed the superiority of the combined score over the GARCE score. CONCLUSIONS Inflammatory biomarkers may play a significant role in the pathophysiology of acute myocardial infarction (AMI) and AMI-related mortality and might be a promising starting point for personalized medicine, which aims to provide each patient with tailored therapy.
Collapse
Affiliation(s)
- T. Schmitz
- grid.419801.50000 0000 9312 0220Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156 Augsburg, Germany
| | - E. Harmel
- grid.419801.50000 0000 9312 0220Department of Cardiology, Respiratory Medicine and Intensive Care, University Hospital Augsburg, Augsburg, Germany
| | - M. Heier
- grid.419801.50000 0000 9312 0220University Hospital of Augsburg, KORA Study Centre, Augsburg, Germany ,Helmholtz Zentrum München, Institute for Epidemiology, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - A. Peters
- Helmholtz Zentrum München, Institute for Epidemiology, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany ,grid.5252.00000 0004 1936 973XChair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany ,grid.452396.f0000 0004 5937 5237German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - J. Linseisen
- grid.419801.50000 0000 9312 0220Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156 Augsburg, Germany
| | - C. Meisinger
- grid.419801.50000 0000 9312 0220Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156 Augsburg, Germany
| |
Collapse
|
29
|
Bo Z, Huang S, Li L, Chen L, Chen P, Luo X, Shi F, Zhu B, Shen L. EGR2 is a hub-gene in myocardial infarction and aggravates inflammation and apoptosis in hypoxia-induced cardiomyocytes. BMC Cardiovasc Disord 2022; 22:373. [PMID: 35971091 PMCID: PMC9377070 DOI: 10.1186/s12872-022-02814-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022] Open
Abstract
Background Myocardial infarction (MI) is characterized by coronary artery occlusion, ischemia and hypoxia of myocardial cells, leading to irreversible myocardial damage. Therefore, it is urgent to explore the potential mechanism of myocardial injury during the MI process to develop effective therapies for myocardial cell rescue. Methods We downloaded the GSE71906 dataset from GEO DataSets, and the R software was used to identify the differentially expressed genes (DEGs) in mouse heart tissues of MI and sham controls. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were performed to understand the significantly activated signaling pathways in MI. Protein–protein interaction (PPI) network was constructed to highlight the hub genes in DEGs. The Western Blot, qRT-PCR and TUNEL staining were used to explore the function of hub gene in hypoxia-induced cardiomyocytes in vitro. Results A total of 235 DEGs were identified in GSE71906 dataset. Functional enrichment analysis revealed that the upregulated genes were primarily associated with the inflammatory response and apoptosis. 20 hub genes were identified in PPI network, and the early growth response 2 (EGR2) was highlighted. In vitro. We confirmed the EGR2 was upregulated induced by hypoxia and revealed the upregulated EGR2 aggravates pro-inflammation and pro-apoptotic genes expression. In addition, EGR2 knockout mitigates hypoxia-induced inflammation and apoptosis in cardiomyocytes. Conclusion The present study identified the EGR2 was a hub gene in myocardial damage during MI process, the excessive EGR2 aggravates hypoxia-induced myocardial damage by accelerating inflammation and apoptosis in vitro. Therefore, targeting EGR2 offers a potential pharmacological strategy for myocardial cell rescue in MI. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02814-3.
Collapse
Affiliation(s)
- Zhixiang Bo
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, #76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Shuwen Huang
- Research Base of Traditional Chinese Medicine Syndrome, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Li Li
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, #76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Lin Chen
- Department of Surgery, Wushan County Hospital of Traditional Chinese Medicine, Chongqing, 400010, China
| | - Ping Chen
- Department of Gastroenterology, The Fifth People's Hospital of Chongqing, Chongqing, 400010, China
| | - Xiaoyi Luo
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, #76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Fang Shi
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, #76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Bing Zhu
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, #76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lin Shen
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, #76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
30
|
Xiao Y, Chen Y, Shao C, Wang Y, Hu S, Lei W. Strategies to improve the therapeutic effect of pluripotent stem cell-derived cardiomyocytes on myocardial infarction. Front Bioeng Biotechnol 2022; 10:973496. [PMID: 35992358 PMCID: PMC9388750 DOI: 10.3389/fbioe.2022.973496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Myocardial infarction (MI) is a common cardiovascular disease caused by permanent loss of cardiomyocytes and the formation of scar tissue due to myocardial ischemia. Mammalian cardiomyocytes lose their ability to proliferate almost completely in adulthood and are unable to repair the damage caused by MI. Therefore, transplantation of exogenous cells into the injured area for treatment becomes a promising strategy. Pluripotent stem cells (PSCs) have the ability to proliferate and differentiate into various cellular populations indefinitely, and pluripotent stem cell-derived cardiomyocytes (PSC-CMs) transplanted into areas of injury can compensate for part of the injuries and are considered to be one of the most promising sources for cell replacement therapy. However, the low transplantation rate and survival rate of currently transplanted PSC-CMs limit their ability to treat MI. This article focuses on the strategies of current research for improving the therapeutic efficacy of PSC-CMs, aiming to provide some inspiration and ideas for subsequent researchers to further enhance the transplantation rate and survival rate of PSC-CMs and ultimately improve cardiac function.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chunlai Shao
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- *Correspondence: Wei Lei, ; Shijun Hu,
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- *Correspondence: Wei Lei, ; Shijun Hu,
| |
Collapse
|
31
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
32
|
Gpx3 and Egr1 Are Involved in Regulating the Differentiation Fate of Cardiac Fibroblasts under Pressure Overload. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3235250. [PMID: 35799890 PMCID: PMC9256463 DOI: 10.1155/2022/3235250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/21/2022] [Accepted: 06/03/2022] [Indexed: 12/04/2022]
Abstract
Objectives Although myocardial fibrosis is a common pathophysiological process associated with many heart diseases, the molecular mechanisms regulating the development of fibrosis have not been fully determined. Recently, single cell RNA sequencing (scRNA-seq) analysis has been used to examine cellular fate and function during cellular differentiation and has contributed to elucidating the mechanisms of various diseases. The main purpose of this study was to characterize the fate of cardiac fibroblasts (CFs) and the dynamic gene expression patterns in a model of cardiac pressure overload using scRNA-seq analysis. Methods The public scRNA-seq dataset of the transverse aortic coarctation (TAC) model in mice was downloaded from the GEO database, GSE155882. First, we performed quality control, dimensionality reduction, clustering, and annotation of the data through the Seurat R package (v4.0.5). Then, we constructed the pseudotime trajectory of cell development and identified key regulatory genes using the Monocle R package (v2.22.0). Different cell fates and groups were fully characterized by Gene Set Enrichment Analysis (GSEA) analysis and Transcription factor (TF) activity analysis. Finally, we used Cytoscape (3.9.1) to extensively examine the gene regulatory network related to cell fate. Results Pseudotime analysis showed that CFs differentiated into two distinct cell fates, one of which produced activated myofibroblasts, and the other which produced protective cells that were associated with reduced fibrosis levels, increased antioxidative stress responses, and the ability to promote angiogenesis. In the TAC model, activated CFs were significantly upregulated, while protective cells were downregulated. Treatment with the bromodomain inhibitor JQ1 reversed this change and improved fibrosis. Analysis of dynamic gene expression revealed that Gpx3 was significantly upregulated during cell differentiation into protective cells. Gpx3 expression was affected by JQ1 treatment. Furthermore, Gpx3 expression levels were negatively correlated with the different levels of fibrosis observed in the various treatment groups. Finally, we found that transcription factors Jun, Fos, Atf3, and Egr1 were upregulated in protective cells, especially Egr1 was predicted to be involved in the regulation of genes related to antioxidant stress and angiogenesis, suggesting a role in promoting differentiation into this cell phenotype. Conclusions The scRNA-seq analysis was used to characterize the dynamic changes associated with fibroblast differentiation and identified Gpx3 as a factor that might be involved in the regulation of myocardial fibrosis under cardiac pressure overload. These findings will help to further understanding of the mechanism of fibrosis and provide potential intervention targets.
Collapse
|
33
|
Research Progress of Fibroblast Growth Factor 21 in Fibrotic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5042762. [PMID: 35677107 PMCID: PMC9168133 DOI: 10.1155/2022/5042762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
Abstract
Fibrosis is a common pathological outcome of chronic injuries, characterized by excessive deposition of extracellular matrix components in organs, as seen in most chronic inflammatory diseases. At present, there is an increasing tendency of the morbidity and mortality of diseases caused by fibrosis, but the treatment measures for fibrosis are still limited. Fibroblast growth factor 21 (FGF21) belongs to the FGF19 subfamily, which also has the name endocrine FGFs because of their endocrine manner. In recent years, it has been found that plasma FGF21 level is significantly correlated with fibrosis progression. Furthermore, there is evidence that FGF21 has a pronounced antifibrotic effect in a variety of fibrotic diseases. This review summarizes the biological effects of FGF21 and discusses what is currently known about this factor and fibrosis disease, highlighting emerging insights that warrant further research.
Collapse
|
34
|
Yang JCS, Huang LH, Wu SC, Wu YC, Wu CJ, Lin CW, Tsai PY, Chien PC, Hsieh CH. Recovery of Dysregulated Genes in Cancer-Related Lower Limb Lymphedema After Supermicrosurgical Lymphaticovenous Anastomosis – A Prospective Longitudinal Cohort Study. J Inflamm Res 2022; 15:761-773. [PMID: 35153500 PMCID: PMC8824698 DOI: 10.2147/jir.s350421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/15/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Johnson Chia-Shen Yang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Lien-Hung Huang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Chan Wu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chia-Jung Wu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chia-Wei Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Pei-Yu Tsai
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Peng-Chen Chien
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ching-Hua Hsieh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Correspondence: Ching-Hua Hsieh, Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, 123 Dapi Road, Niaosong District, Kaohsiung City, 833, Taiwan, Tel +886-7-7317123, ext.8002, Fax +886-7-7354309, Email
| |
Collapse
|