1
|
Fan Y, Jialiken D, Zheng Z, Zhang W, Zhang S, Zheng Y, Sun Z, Zhang H, Yan X, Liu M, Fang Z. Qianyang Yuyin granules alleviate hypertension-induced vascular remodeling by inhibiting the phenotypic switch of vascular smooth muscle cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118896. [PMID: 39393558 DOI: 10.1016/j.jep.2024.118896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/21/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qianyang Yuyin granules (QYYY) have been used clinically to treat hypertension for over two decades. Previous clinical trials have shown that QYYY can improve vascular elastic function in hypertensive patients. However, the underlying pharmacological mechanism is unclear. AIM OF THE STUDY To elucidate the effects and mechanisms of QYYY on vascular remodeling using a multidisciplinary approach that includes network pharmacology, proteomics, and both in vitro and in vivo experiments. MATERIALS AND METHODS The main components of QYYY were identified using ultra-high-performance liquid chromatography and high-resolution mass spectrometry. Network pharmacology and molecular docking were employed to predict QYYY's primary active ingredients, potential therapeutic targets and intervention pathways in hypertensive vascular remodeling. We induced hypertension in male C57BL/6 mice by infusing angiotensin II (Ang II) via osmotic minipumps, and performed pre-treatment with QYYY or Sacubitril/valsartan (Entresto). Blood pressure was monitored in vivo, followed by the extraction of aortas to examine pathological structural changes and alterations in protein expression patterns. The expression and location of proteins involved in the HIF-1α/TWIST1/P-p65 signaling pathway were investigated, as well as markers of vascular smooth muscle cells (VSMCs) phenotypic switch. In vitro, we studied the effects of QYYY water extract on Ang II-stimulated human aortic VSMCs. We investigated whether QYYY could affect the HIF-1α/TWIST1/P-p65 signaling pathway, thereby ameliorating apoptosis, autophagy, and phenotype switch in VSMCs. RESULTS We identified 62 main compounds in QYYY, combined with network pharmacology, speculated 827 potentially active substances, and explored 1021 therapeutic targets. The KEGG pathway analysis revealed that the mechanisms of action associated with QYYY therapy potentially encompass various biological processes, including metabolic pathways, TNF signaling pathways, apoptosis, Ras signaling pathways, HIF-1 signaling pathways, autophagy-animal pathways. In hypertensive mice, QYYY restored abnormally elevated blood pressure, vascular remodeling, and inflammation with a dose-response relationship while altering abnormal protein patterns. In vitro, QYYY could inhibit abnormal proliferation, migration, intracellular Ca2+ accumulation and cytoskeletal changes of VSMCs. It improved mitochondrial function, reduced ROS levels, stabilized membrane potential, prevented cell death, and reduced overproduction of TGF-β1, TNF-a, and IL-1β. CONCLUSION QYYY may be able to inhibit the overactivation of the HIF-1α/TWIST1/P-p65 signaling pathway, improve the phenotypic switch, and balance apoptosis and autophagy in VSMCs, thereby effectively improving vascular remodeling caused by hypertension.
Collapse
Affiliation(s)
- Yadong Fan
- Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China.
| | - Dinala Jialiken
- Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ziwen Zheng
- Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Weiting Zhang
- Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Siqi Zhang
- Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Yawei Zheng
- Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zeqi Sun
- Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Haitao Zhang
- Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiwu Yan
- Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China.
| | - Ming Liu
- Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Zhuyuan Fang
- Institute of Hypertension, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Jiangsu Chinese Medicine Clinical Medicine Innovation Center for Hypertension, Nanjing, 210029, China; Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
2
|
Aristizábal-Colorado D, Ocampo-Posada M, Rivera-Martínez WA, Corredor-Rengifo D, Rico-Fontalvo J, Gómez-Mesa JE, Duque-Ossman JJ, Abreu-Lomba A. SGLT2 Inhibitors and How They Work Beyond the Glucosuric Effect. State of the Art. Am J Cardiovasc Drugs 2024; 24:707-718. [PMID: 39179723 DOI: 10.1007/s40256-024-00673-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 08/26/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with a heightened risk of cardiovascular and renal complications. While glycemic control remains essential, newer therapeutic options, such as SGLT2 inhibitors, offer additional benefits beyond glucose reduction. This review delves into the mechanisms underlying the cardio-renal protective effects of SGLT2 inhibitors. By inducing relative hypoglycemia, these agents promote ketogenesis, optimize myocardial energy metabolism, and reduce lipotoxicity. Additionally, SGLT2 inhibitors exert renoprotective actions by enhancing renal perfusion, attenuating inflammation, and improving iron metabolism. These pleiotropic effects, including modulation of blood pressure, reduction of uric acid, and improved endothelial function, collectively contribute to the cardiovascular and renal benefits observed with SGLT2 inhibitor therapy. This review will provide clinicians with essential knowledge, understanding, and a clear recollection of this pharmacological group's mechanism of action.
Collapse
Affiliation(s)
- David Aristizábal-Colorado
- Department of Internal Medicine, Universidad Libre, Cali, Colombia
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Interamerican Society of Cardiology (SIAC), Mexico City, Mexico
| | - Martín Ocampo-Posada
- Department of Internal Medicine, Universidad Libre, Cali, Colombia
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Faculty of Health, Pontificia Universidad Javeriana, Cali, Colombia
- Grupo de Investigación en Ciencias Básicas y Clínicas de la Salud, Universidad Javeriana, Cali, Colombia
| | - Wilfredo Antonio Rivera-Martínez
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Department of Endocrinology, Faculty of Medicine, Universidad de Antioquia, Medellin, Colombia
| | - David Corredor-Rengifo
- Department of Internal Medicine, Universidad Libre, Cali, Colombia
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
| | - Jorge Rico-Fontalvo
- Department of Nephrology. Faculty of Medicine, Universidad Simón Bolívar, Barranquilla, Colombia
- Latin American Society of Nephrology and Arterial Hypertension (SLANH), Panama City, Panamá
| | - Juan Esteban Gómez-Mesa
- Interamerican Society of Cardiology (SIAC), Mexico City, Mexico.
- Cardiology Department, Fundación Valle del Lili, Cali, Colombia.
- Department of Health Sciences, Universidad Icesi, Cali, Colombia.
| | - John Jairo Duque-Ossman
- Universidad Del Quindío, Armenia, Colombia
- Latin American Federation of Endocrinology (FELAEN), Armenia, Colombia
| | - Alin Abreu-Lomba
- Internal Medicine Research Group, Universidad Libre, Cali, Colombia
- Endocrinology Department, Clínica Imbanaco, Cali, Colombia
| |
Collapse
|
3
|
Zhang Z, Wu H, Zhang A, Tan M, Yan S, Jiang D. Transfer of heavy metals along the food chain: A review on the pest control performance of insect natural enemies under heavy metal stress. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135587. [PMID: 39186843 DOI: 10.1016/j.jhazmat.2024.135587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Heavy metal contamination represents a critical global environmental concern. The movement of heavy metals through the food chain inevitably subjects insect natural enemies to heavy metal stress, leading to various adverse effects. This review assesses the risks posed by heavy metal exposure to insect natural enemies, evaluates how such exposure impacts their pest control efficacy, and investigates the mechanisms affecting their fitness. Heavy metals transfer and accumulate from soil to plants, then to herbivorous insects, and ultimately to their natural enemies, impeding growth, development, and reproduction of insect natural enemies. Typically, diminished growth and reproduction directly compromise the pest control efficacy of these natural enemies. Nonetheless, within tolerable limits, increased feeding may occur as these natural enemies strive to meet the energy demands for detoxification, potentially enhancing their pest control capabilities. The production of reactive oxygen species and oxidative damage caused by heavy metals in insect natural enemies, combined with disrupted energy metabolism in host insects, are key factors contributing to the reduced fitness of insect natural enemies. In summary, heavy metal pollution emerges as a significant abiotic factor adversely impacting the pest control performance of these beneficial insects.
Collapse
Affiliation(s)
- Zhe Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Hongfei Wu
- School of Forestry, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China; Forest conservation institute, Chinese Academy of Forestry, Harbin 150040, China
| | - Aoying Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Mingtao Tan
- School of Forestry, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Shanchun Yan
- School of Forestry, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Dun Jiang
- School of Forestry, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
4
|
Gong X, Zhao Q, Zhang H, Liu R, Wu J, Zhang N, Zou Y, Zhao W, Huo R, Cui R. The Effects of Mesenchymal Stem Cells-Derived Exosomes on Metabolic Reprogramming in Scar Formation and Wound Healing. Int J Nanomedicine 2024; 19:9871-9887. [PMID: 39345908 PMCID: PMC11438468 DOI: 10.2147/ijn.s480901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Pathological scarring results from aberrant cutaneous wound healing due to the overactivation of biological behaviors of human skin fibroblasts, characterized by local inordinate inflammation, excessive extracellular matrix and collagen deposition. Yet, its underlying pathogenesis opinions vary, which could be caused by increased local mechanical tension, enhanced and continuous inflammation, gene mutation, as well as cellular metabolic disorder, etc. Metabolic reprogramming is the process by which the metabolic pattern of cells undergoes a systematic adjustment and transformation to adapt to the changes of the external environment and meet the needs of their growth and differentiation. Therefore, the abnormality of metabolic reprogramming in cells within wounds and scars attaches great importance to scar formation. Mesenchymal stem cells-derived exosomes (MSC-Exo) are the extracellular vesicles that play an important role in tissue repair, cancer treatment as well as immune and metabolic regulation. However, there is not a systematic work to detail the relevant studies. Herein, we gave a comprehensive summary of the existing research on three main metabolisms, including glycometabolism, lipid metabolism and amino acid metabolism, and MSC-Exo regulating metabolic reprogramming in wound healing and scar formation for further research reference.
Collapse
Affiliation(s)
- Xiangan Gong
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Qian Zhao
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Huimin Zhang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Rui Liu
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Jie Wu
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Nanxin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, People’s Republic of China
| | - Yuanxian Zou
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Wen Zhao
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Ran Huo
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Rongtao Cui
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, People’s Republic of China
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
5
|
Xiang T, Yang C, Deng Z, Sun D, Luo F, Chen Y. Krüppel-like factors family in health and disease. MedComm (Beijing) 2024; 5:e723. [PMID: 39263604 PMCID: PMC11387732 DOI: 10.1002/mco2.723] [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: 03/19/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Krüppel-like factors (KLFs) are a family of basic transcription factors with three conserved Cys2/His2 zinc finger domains located in their C-terminal regions. It is acknowledged that KLFs exert complicated effects on cell proliferation, differentiation, survival, and responses to stimuli. Dysregulation of KLFs is associated with a range of diseases including cardiovascular disorders, metabolic diseases, autoimmune conditions, cancer, and neurodegenerative diseases. Their multidimensional roles in modulating critical pathways underscore the significance in both physiological and pathological contexts. Recent research also emphasizes their crucial involvement and complex interplay in the skeletal system. Despite the substantial progress in understanding KLFs and their roles in various cellular processes, several research gaps remain. Here, we elucidated the multifaceted capabilities of KLFs on body health and diseases via various compliable signaling pathways. The associations between KLFs and cellular energy metabolism and epigenetic modification during bone reconstruction have also been summarized. This review helps us better understand the coupling effects and their pivotal functions in multiple systems and detailed mechanisms of bone remodeling and develop potential therapeutic strategies for the clinical treatment of pathological diseases by targeting the KLF family.
Collapse
Affiliation(s)
- Tingwen Xiang
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Chuan Yang
- Department of Biomedical Materials Science Third Military Medical University (Army Medical University) Chongqing China
| | - Zihan Deng
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Dong Sun
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Fei Luo
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Yueqi Chen
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
- Department of Orthopedics Chinese PLA 76th Army Corps Hospital Xining China
| |
Collapse
|
6
|
Li L, Ye J, Zhao Z, Hu S, Liang H, Ouyang J, Hu Z. Shenfu injection improves isoproterenol-induced heart failure in rats by modulating co-metabolism and regulating the trimethylamine-N-oxide - inflammation axis. Front Pharmacol 2024; 15:1412300. [PMID: 38966553 PMCID: PMC11222397 DOI: 10.3389/fphar.2024.1412300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024] Open
Abstract
Heart failure (HF) is a chronic condition that progressively worsens and continues to be a major financial burden and public health concern. The "gut-heart" axis provides an innovative perspective and therapeutic strategy for preventing and treating heart failure. Shenfu injection (SFI) is a Traditional Chinese Medicine-based treatment demonstrating potential as a therapeutic strategy for heart failure. However, the precise therapeutic mechanisms of SFI in heart failure are not completely characterized. In this study, HF models were established utilizing subcutaneous multipoint injection of isoproterenol (ISO) at a dosage of 5 mg kg-1·d-1 for 7 days. Serum levels of inflammatory biomarkers were quantified using protein microarrays. Rat feces were analyzed using untargeted metabolomics research and 16S rRNA sequencing. The link between gut microbiota and metabolites was examined using a MetOrigin and Spearman correlation analysis. Our results show that Shenfu injection effectively enhances cardiac function in rats with ISO-induced heart failure by potentially modulating pro-/anti-inflammatory imbalance and reducing serum and urine Trimethylamine-N-oxide (TMAO) levels. Moreover, SFI significantly increases the abundance of Bacteroidota at the phylum level, thereby improving disrupted gut microbiota composition. Additionally, SFI supplementation enriches specific genera known for their capacity to produce short-chain fatty acids. SFI was found to be associated with three key metabolic pathways, as revealed by fecal metabonomics analysis, including the pentose phosphate pathway, pyrimidine metabolism, and purine metabolism. Metabolite tracing analysis revealed that Taurine and hypotaurine metabolism was found to be specific to the microbial community. The biosynthesis of Pyrimidine metabolism, Purine metabolism, beta-alanine metabolism, Naphthalene degradation, Pantothenate, and CoA biosynthesis were identified as co-metabolic pathways between microbes and host. The Spearman correlation analysis was also significantly correlated to differentially expressed metabolites regulated by SFI and the gut microbiota. These results suggest that SFI improves ISO-induced heart failure by modulating co-metabolism and regulating the TMAO-inflammation axis.
Collapse
Affiliation(s)
- Lin Li
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiahao Ye
- Post-Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhenyu Zhao
- Post-Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyuan Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hao Liang
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ji Ouyang
- Post-Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhixi Hu
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
7
|
Hu YX, Qiu SL, Shang JJ, Wang Z, Lai XL. Pharmacological Effects of Botanical Drugs on Myocardial Metabolism in Chronic Heart Failure. Chin J Integr Med 2024; 30:458-467. [PMID: 37750985 DOI: 10.1007/s11655-023-3649-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 09/27/2023]
Abstract
Although there have been significant advances in the treatment of heart failure in recent years, chronic heart failure remains a leading cause of cardiovascular disease-related death. Many studies have found that targeted cardiac metabolic remodeling has good potential for the treatment of heart failure. However, most of the drugs that increase cardiac energy are still in the theoretical or testing stage. Some research has found that botanical drugs not only increase myocardial energy metabolism through multiple targets but also have the potential to restore the balance of myocardial substrate metabolism. In this review, we summarized the mechanisms by which botanical drugs (the active ingredients/formulas/Chinese patent medicines) improve substrate utilization and promote myocardial energy metabolism by activating AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptors (PPARs) and other related targets. At the same time, some potential protective effects of botanical drugs on myocardium, such as alleviating oxidative stress and dysbiosis signaling, caused by metabolic disorders, were briefly discussed.
Collapse
Affiliation(s)
- Yu-Xuan Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Sheng-Lei Qiu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Ju-Ju Shang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Zi Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Xiao-Lei Lai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| |
Collapse
|
8
|
Cheng L, Liang J, Xie F, Han Z, Luo W, Chen H, He J. Identification and validation of a novel glycolysis-related ceRNA network for sepsis-induced cardiomyopathy. Front Med (Lausanne) 2024; 11:1343281. [PMID: 38439898 PMCID: PMC10910075 DOI: 10.3389/fmed.2024.1343281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Purpose Sepsis-induced cardiomyopathy (SIC) is a major life-threatening condition in critically infected patients. Early diagnosis and intervention are important to improve patient prognosis. Recognizing the pivotal involvement of the glycolytic pathway in SIC, this study aims to establish a glycolysis-related ceRNA network and explore novel diagnostic avenues. Materials and methods SIC-related datasets were carefully filtered from the GEO database. CytoHubba was used to identify differentially expressed genes (DEGs) associated with glycolysis. A predictive method was then used to construct an lncRNA-miRNA-mRNA network. Dual-luciferase reporter assays validated gene interactions, and the specificity of this ceRNA network was confirmed in peripheral blood mononuclear cells (PBMCs) from SIC patients. Logistic analysis was used to examine the correlation between the ceRNA network and SIC. Diagnostic potential was assessed using receiver operating characteristic (ROC) curves, and correlation analysis investigated any associations between gene expression and clinical indicators. Results IER3 was identified as glycolysis-related DEG in SIC, and a ceRNA network (SNHG17/miR-214-3p/IER3) was established by prediction. Dual luciferase reporter gene assay confirmed the presence of mutual binding between IER3, miR-214-3p and SNHG17. RT-qPCR verified the specific expression of this ceRNA network in SIC patients. Multivariate logistic analysis established the correlation between the ceRNA network and SIC. ROC analysis demonstrated its high diagnostic specificity (AUC > 0.8). Correlation analysis revealed a negative association between IER3 expression and oxygenation index in SIC patients (p < 0.05). Furthermore, miR-214-3p expression showed a negative correlation with NT-proBNP (p < 0.05). Conclusion In this study, we identified and validated a ceRNA network associated with glycolysis in SIC: SNHG17/miR-214-3p/IER3. This ceRNA network may play a critical role in the onset and development of SIC. This finding is important to further our understanding of the pathophysiological mechanisms underlying SIC and to explore potential diagnostic and therapeutic targets for SIC.
Collapse
Affiliation(s)
- Lulu Cheng
- Postgraduate Cultivation Base of Guangzhou University of Chinese Medicine, Panyu Central Hospital, Guangzhou, China
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jiabin Liang
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Zeping Han
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Hanwei Chen
- Radiology Department of Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou, China
| | - Jinhua He
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| |
Collapse
|
9
|
Packer M. Hyperuricemia and Gout Reduction by SGLT2 Inhibitors in Diabetes and Heart Failure: JACC Review Topic of the Week. J Am Coll Cardiol 2024; 83:371-381. [PMID: 38199714 DOI: 10.1016/j.jacc.2023.10.030] [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: 08/21/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 01/12/2024]
Abstract
Gout is characterized by increased production of purines (through the pentose phosphate pathway), which is coupled with reduced renal or intestinal excretion of urate. Concurrent upregulation of nutrient surplus signaling (mammalian target of rapamycin and hypoxia-inducible factor-1a) and downregulation of nutrient deprivation signaling (sirtuin-1 and adenosine monophosphate-activated protein kinase) redirects glucose toward anabolic pathways (rather than adenosine triphosphate production), thus promoting heightened oxidative stress and cardiomyocyte and proximal tubular dysfunction, leading to cardiomyopathy and kidney disease. Hyperuricemia is a marker (rather than a driver) of these cellular stresses. By inducing a state of starvation mimicry in a state of nutrient surplus, sodium-glucose cotransporter-2 inhibitors decrease flux through the pentose phosphate pathway (thereby attenuating purine and urate synthesis) while promoting renal urate excretion. These convergent actions exert a meaningful effect to lower serum uric acid by ≈0.6 to 1.5 mg/dL and to reduce the risk of gout by 30% to 50% in large-scale clinical trials.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, Texas, USA; Imperial College, London, United Kingdom.
| |
Collapse
|
10
|
Wang L, Han Y, Gu Z, Han M, Hu C, Li Z. Boosting the therapy of glutamine-addiction glioblastoma by combining glutamine metabolism therapy with photo-enhanced chemodynamic therapy. Biomater Sci 2023; 11:6252-6266. [PMID: 37534821 DOI: 10.1039/d3bm00897e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
The complete treatment of high grade invasive glioblastoma (GBM) remains to be a great challenge, and it is of great importance to develop innovative therapeutic approaches. Herein, we found that GBM derived from U87 MG cells is a glutamine-addiction tumor, and jointly using glutamine-starvation therapy and photo-enhanced chemodynamic therapy (CDT) can significantly boost its therapy. We rationally fabricated tumor cell membrane coated Cu2-xSe nanoparticles (CS NPs) and an inhibitor of glutamine metabolism (Purpurin) for combined therapy, because glutamine rather than glucose plays a crucial role in the proliferation and growth of GBM cells, and serves as a precursor for the synthesis of glutathione (GSH). The resultant CS-P@CM NPs can be specifically delivered to the tumor site to inhibit glutamine metabolism in tumor cells, suppress tumor intracellular GSH, and increase H2O2 content, which benefit the CDT catalyzed by CS NPs. The cascade reaction can be further enhanced by irradiation with the second near-infrared (NIR-II) light at the maximum concentration of H2O2, which can be monitored by photoacoustic imaging. The NIR-II light irradiation can generate a large amount of reactive oxygen species (ROS) within a short time to kill tumor cells and enhance the CDT efficacy. This is the first work on the treatment of orthotopic malignant GBM through combined glutamine metabolism therapy and photo-enhanced CDT, and provides insights into the treatment of other solid tumors by modulating the metabolism of tumor cells.
Collapse
Affiliation(s)
- Ling Wang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Zhengpeng Gu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|