1
|
Pan Y, Chen B, Xie J, Chen D, Cai Y, Zhao D, Cao Y, Lian F, Yan X. Lentinan alleviates angiotensin II-induced myocardial remodeling through LMP7-SOCS3 signaling. Int J Biol Macromol 2025:142146. [PMID: 40101827 DOI: 10.1016/j.ijbiomac.2025.142146] [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: 11/21/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Myocardial remodeling is a major pathological mechanism causing heart failure. As a critical negative modulator of cardiac remodeling, suppressor of cytokine signaling 3 (SOCS3) is regulated by immunoproteasome subunit large multifunctional peptidase 7 (LMP7). Lentinan (LNT), a β-polysaccharide extracted from Lentinus edodes, has anti-inflammatory and antioxidant properties. However, the role and molecular mechanisms of LNT in angiotensin II (Ang II)-triggered myocardial remodeling are unclear. Myocardial remodeling was established using Ang II infusion (1000 or 200 ng/kg/min) for 2 weeks. Cardiomyocytes and cardiac fibroblasts were triggered by Ang II. LNT was administered daily by oral gavage to mice starting 1 day before Ang II or saline treatment. Here, we found that LNT supplementation dose-dependently ameliorated Ang II-triggered myocardial dysfunction and remodeling (hypertrophy, fibrosis, inflammation, and superoxide production). Mechanistically, LNT suppressed SOCS3 protein degradation by downregulating immunoproteasome LMP7 activity and expression, thereby inactivating downstream signaling, such as STAT3, ERK, AKT, NF-κB, and TGF-β. Conversely, SOCS3 knockdown significantly blocked the protective effect of LNT on myocardial remodeling in Ang II-infused mice. Together, our findings suggest that LNT may be a new therapeutic approach for myocardial remodeling and heart failure.
Collapse
Affiliation(s)
- Yu Pan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Bingqi Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiawen Xie
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Danni Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuwei Cai
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Denghui Zhao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yifei Cao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Fuzhi Lian
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China; Engineering Research Center of Mobile Health Management System, Ministry of Education, Hangzhou, China
| | - Xiao Yan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
2
|
Taboada-Alquerque M, Olivero-Verbel J. Network Toxicology Analysis Reveals Molecular Mechanisms Associated with Noise Exposure to Multiple Diseases. Toxicol Mech Methods 2025:1-25. [PMID: 39898607 DOI: 10.1080/15376516.2025.2460591] [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: 05/02/2024] [Revised: 12/09/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Noise pollution is recognized as an environmental stressor that affects various biological processes beyond auditory functions, mainly through stress hormones release. This work explored the biological processes, diseases attributable to noise-regulated targets, and the main targets involved in each disease, employing a network toxicology approach. Through various databases and bioinformatics analysis, a total of 577 targets were identified as potential candidates implicated in diseases related to noise exposure, 10 from the GEO database and the rest from other databases. Noise pollution was found to regulate processes such as hormone response, cellular response to cytokines, and circulatory system functions, contributing to the development of the pathological manifestations related to the diseases like hypertension, ischemia, atherosclerosis, and cirrhosis. Hub targets for ischemia included IL-6, CASP3, AKT1, and TNF-α, while NOS3 was related to hypertension, and NOS3, TNF-α, AGT, and IL-1B to atherosclerosis. The targets were found to be linked to vascular regulation and inflammation in cardio- and cerebrovascular diseases. Molecular docking studies indicated stress hormones released by noise exposure regulates these diseases through signaling pathways, without implicating its direct binding to hub targets. The results indicate that individuals with vascular diseases are more vulnerable to the effects of prolonged noise exposure.
Collapse
Affiliation(s)
- Maria Taboada-Alquerque
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, Zaragocilla Campus, University of Cartagena, Cartagena 130014, Colombia
| | - Jesus Olivero-Verbel
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, Zaragocilla Campus, University of Cartagena, Cartagena 130014, Colombia
| |
Collapse
|
3
|
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
|
4
|
Zhao XL, Cao ZJ, Li KD, Tang F, Xu LY, Zhang JN, Liu D, Peng C, Ao H. Gallic acid: a dietary metabolite's therapeutic potential in the management of atherosclerotic cardiovascular disease. Front Pharmacol 2025; 15:1515172. [PMID: 39840111 PMCID: PMC11747375 DOI: 10.3389/fphar.2024.1515172] [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: 10/22/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) causes significant morbidity and mortality globally. Most of the chemicals specifically target certain pathways and minimally impact other diseases associated with ASCVD. Moreover, interactions of these drugs can cause toxic reactions. Consequently, the exploration of multi-targeted and safe medications for treating and preventing ASCVD has become an increasingly popular trend. Gallic acid (GA), a natural secondary metabolite found in various fruits, plants, and nuts, has demonstrated potentials in preventing and treating ASCVD, in addition to its known antioxidant and anti-inflammatory effects. It alleviates the entire process of atherosclerosis (AS) by reducing oxidative stress, improving endothelial dysfunction, and inhibiting platelet activation and aggregation. Additionally, GA can treat ASCVD-related diseases, such as coronary heart disease (CHD) and cerebral ischemia. However, the pharmacological actions of GA in the prevention and treatment of ASCVD have not been comprehensively reviewed, which limits its clinical development. This review primarily summarizes the in vitro and in vivo pharmacological actions of GA on the related risk factors of ASCVD, AS, and ASCVD. Additionally, it provides a comprehensive overview of the toxicity, extraction, synthesis, pharmacokinetics, and pharmaceutics of GA,aimed to enhance understanding of its clinical applications and further research and development.
Collapse
Affiliation(s)
- Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhang-Jing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke-Di Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
Wongwad E, Preedalikit W, Changprasoed S, Somsai S, Singmee N, Srisuksomwong P, Srivilai J, Rungsang T, Mungmai L. Effects of Different Drying Processes on the Bioactivity and Rutin Content of Prunus spp. (Plums). INTERNATIONAL JOURNAL OF FOOD SCIENCE 2024; 2024:9999731. [PMID: 39583586 PMCID: PMC11585372 DOI: 10.1155/2024/9999731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/23/2024] [Indexed: 11/26/2024]
Abstract
The aim of this study was to investigate the effects of drying process preparations, solvent type, and different species of Prunus spp. (European, Japanese, and cherry plums) on bioactive properties (antioxidant, anti-lipid peroxidation, anti-tyrosinase, and anti-inflammatory activities) and rutin content. Leaves and fresh fruits of plums were dried using a cold process (vacuum freeze dryer at -105 ± 5°C) and a hot process (hot air oven at 50 ± 0.5°C). The dried plant material was then extracted using ethanol and propylene glycol to obtain ethanolic crude extracts and propylene glycol extract solutions. These extracts were then tested for phenolic and flavonoid contents and their potential biological activities and analyzed for rutin content using the HPLC method. The results showed that the ethanolic extract of plum leaves obtained via the cold-drying process exhibited higher total phenolic and flavonoid contents (96.94 ± 6.73-112.34 ± 9.08 mg GAE/g sample and 105.10 ± 11.31-185.94 ± 23.35 mg QE/g sample, respectively), as well as greater potential for antioxidant activity (DPPH with IC50 = 29.97 ± 1.17 to 31.44 ± 4.16 μg/mL), anti-tyrosinase activity (40.34 ± 1.27%-46.91 ± 0.22%), and anti-lipid peroxidation activity (34.10 ± 4.88%-38.27 ± 2.12%). They also exhibited higher anti-inflammatory activity by inhibiting NO (20.16 ± 0.12%-40.05 ± 0.42%), IL-6 (37.81 ± 3.01%-42.37 ± 8.92%), and TNF-α (45.96 ± 7.93%-63.28 ± 6.44%) compared to fresh plum fruit extracts and other extraction procedures. These activities correlated with the rutin content, which was high in the plum leaf extracts (28.52 ± 0.00%-43.82 ± 0.13% w/w). European plums tended to exhibit greater bioactivities and higher rutin content compared to other species. These findings indicate that plum leaf extracts are a promising source for further applications in food, nutrition, and health products.
Collapse
Affiliation(s)
- Eakkaluk Wongwad
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Research and Innovation Center in Cosmetic Sciences and Natural Products, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Weeraya Preedalikit
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Research and Innovation Center in Cosmetic Sciences and Natural Products, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Suttida Changprasoed
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Suthida Somsai
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Nanthawan Singmee
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Pawalee Srisuksomwong
- Division of Science and Mathematics, Faculty of Science and Technology, Phuket Rajabhat University, Phuket 83000, Thailand
| | - Jukkarin Srivilai
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Research and Innovation Center in Cosmetic Sciences and Natural Products, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Tammanoon Rungsang
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Research and Innovation Center in Cosmetic Sciences and Natural Products, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Lapatrada Mungmai
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Research and Innovation Center in Cosmetic Sciences and Natural Products, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| |
Collapse
|
6
|
Othman NS, Nor Hisam NS, Mad Azli AA, Mansor NI, Hamid AA, Aminuddin A, Mohamad Anuar NN, Ahmad MF, Ugusman A. Persicaria minor (Huds.) Opiz Exhibits Antihypertensive Effects by Inhibiting the Angiotensin-Converting Enzyme/Angiotensin II Type 1 Receptor Pathway in Human Endothelial Cells. Life (Basel) 2024; 14:1486. [PMID: 39598284 PMCID: PMC11595449 DOI: 10.3390/life14111486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Overactivation of the angiotensin-converting enzyme (ACE)/angiotensin II type 1 receptor (AT1R) pathway leads to vasoconstriction and elevated blood pressure. Persicaria minor (Huds.) Opiz is an herbal plant known for its antioxidant, anti-hyperlipidemic, and anti-atherosclerotic properties, with bioactive compounds that exhibit antihypertensive effects. Therefore, this study aimed to evaluate the antihypertensive effects of the standardized aqueous extract of P. minor leaf (AEPM) through the ACE/AT1R pathway in human umbilical vein endothelial cells (HUVECs) induced with phorbol 12-myristate 13-acetate (PMA). HUVECs were stimulated with PMA to induce ACE, with or without AEPM or captopril treatment, for 24 h. Subsequently, ACE mRNA expression, ACE protein levels, ACE activity, angiotensin II levels, and AT1R expression were measured. The results demonstrated that AEPM treatment significantly reduced ACE mRNA expression, ACE protein levels, ACE activity, angiotensin II levels, and AT1R expression in PMA-induced HUVECs. The modulatory effects of AEPM on the ACE/AT1R pathway were comparable to those of captopril. Ex vivo experiments further confirmed that AEPM reduced the contraction responses of rat aortic rings to PMA. In conclusion, P. minor effectively inhibits the ACE/AT1R pathway in PMA-induced HUVECs, suggesting its potential as a natural antihypertensive agent.
Collapse
Affiliation(s)
- Nur Syakirah Othman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.S.O.); (N.S.N.H.); (A.A.M.A.); (A.A.H.); (A.A.)
| | - Nur Syahidah Nor Hisam
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.S.O.); (N.S.N.H.); (A.A.M.A.); (A.A.H.); (A.A.)
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Amanina Athirah Mad Azli
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.S.O.); (N.S.N.H.); (A.A.M.A.); (A.A.H.); (A.A.)
| | - Nur Izzati Mansor
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.S.O.); (N.S.N.H.); (A.A.M.A.); (A.A.H.); (A.A.)
- Cardiovascular and Pulmonary Research Group (CardioResp), Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.S.O.); (N.S.N.H.); (A.A.M.A.); (A.A.H.); (A.A.)
- Cardiovascular and Pulmonary Research Group (CardioResp), Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Nur Najmi Mohamad Anuar
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Mohd Faizal Ahmad
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.S.O.); (N.S.N.H.); (A.A.M.A.); (A.A.H.); (A.A.)
- Cardiovascular and Pulmonary Research Group (CardioResp), Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
7
|
Bukowska P, Bralewska M, Pietrucha T, Sakowicz A. Nutraceuticals as Modulators of Molecular Placental Pathways: Their Potential to Prevent and Support the Treatment of Preeclampsia. Int J Mol Sci 2024; 25:12167. [PMID: 39596234 PMCID: PMC11594370 DOI: 10.3390/ijms252212167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Preeclampsia (PE) is a serious condition characterized by new-onset hypertension and proteinuria or organ dysfunction after the 20th week of gestation, making it a leading cause of maternal and fetal mortality worldwide. Despite extensive research, significant gaps remain in understanding the mechanisms underlying PE, contributing to the ineffectiveness of current prevention and treatment strategies. Consequently, premature cesarean sections often become the primary intervention to safeguard maternal and fetal health. Emerging evidence indicates that placental insufficiency, driven by molecular disturbances, plays a central role in the development of PE. Additionally, the maternal microbiome may be implicated in the pathomechanism of preeclampsia by secreting metabolites that influence maternal inflammation and oxidative stress, thereby affecting placental health. Given the limitations of pharmaceuticals during pregnancy due to potential risks to fetal development and concerns about teratogenic effects, nutraceuticals may provide safer alternatives. Nutraceuticals are food products or dietary supplements that offer health benefits beyond basic nutrition, including plant extracts or probiotics. Their historical use in traditional medicine has provided valuable insights into their safety and efficacy, including for pregnant women. This review will examine how the adoption of nutraceuticals can enhance dysregulated placental pathways, potentially offering benefits in the prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
| | | | | | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
8
|
Zhu C, Lin Z, Jiang H, Wei F, Wu Y, Song L. Recent Advances in the Health Benefits of Phenolic Acids in Whole Grains and the Impact of Processing Techniques on Phenolic Acids: A Comprehensive Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24131-24157. [PMID: 39441722 DOI: 10.1021/acs.jafc.4c05245] [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: 10/25/2024]
Abstract
Phenolic acids, essential compounds in whole grains, are renowned for their health-enhancing antioxidant and anti-inflammatory properties. Variations in concentration, particularly of hydroxybenzoic and hydroxycinnamic acids, are observed among grain types. Their antiobesity and antidiabetes effects are linked to their modulation of key signaling pathways like AMPK and PI3K, crucial for metabolic regulation and the body's response to inflammation and oxidative stress. Processing methods significantly influence phenolic acid content and bioavailability in whole grains. Thermal techniques like boiling, baking, or roasting can degrade these compounds, with loss influenced by processing conditions. Nonthermal methods such as germination, fermentation, or their combination, can protect or enhance phenolic acid content under ideal conditions. Novel nonthermal approaches like ultrahigh pressure (UHP), irradiation, and pulsed electric fields (PEF) show promise in preserving these compounds. Further research is needed to fully comprehend the impact mechanisms of these innovative methods on the nutritional and sensory attributes of cereals.
Collapse
Affiliation(s)
- Chuang Zhu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zihan Lin
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huibin Jiang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fenfen Wei
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan Wu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lihua Song
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- Center of Hydrogen Science, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
9
|
Wang S, Zhu C, Zhang S, Ma S, Li B, Zhao S, Zhang W, Sun Z. The Combination of Gastrodin and Gallic Acid Synergistically Attenuates AngII-Induced Apoptosis and Inflammation via Regulation of Sphingolipid Metabolism. J Inflamm Res 2024; 17:6971-6988. [PMID: 39372584 PMCID: PMC11456272 DOI: 10.2147/jir.s477554] [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: 07/10/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024] Open
Abstract
Background Hypertension (HTN) is closely related to endothelial damage. While tianma (TM) and gouqizi (GQZ) have the potential to be effective in the treatment of HTN in traditional Chinese medicine, their main active ingredients and whether its exert synergistic effects and the underlying mechanisms of synergistic effects are still unclear. Objective This study screened the active ingredients of TM and GQZ, investigated the synergistic effects of the active ingredients and explored possible mechanisms. Methods The potential targets and mechanisms of TM and GQZ were screened using network pharmacology, and gastrodin (GAS) and gallic acid (GA) were identified as compounds with significant antihypertensive activity. The synergistic effects of the combination of GAS and GA was assessed by measuring biomarkers of AngII-induced human umbilical vein endothelial cell (HUVECs) dysfunction model. Furthermore, the anti-apoptotic and anti-inflammatory effects were evaluated by measuring inflammatory cytokine secretion, and apoptosis-related markers. Finally, key targets of the sphingolipid signaling pathway were experimentally validated by Western blotting. Results In network pharmacology, the herb-pair exerted a synergetic effect by regulating sphingolipid pathways. The GAS and GA exerted synergistic protective effects in AngII-induced HUVECs injury by improving Nitric Oxide Content (NO) levels, alleviating lactate Endothelin-1 (ET-1), and Thromboxane B2 (TX-B2) release, reducing the secretion of inflammatory factors like interleukin-6 (IL-6), interleukin-1β (IL-1β), Tumor Necrosis Factor Alpha (TNF-α)), decreasing the pro-apoptotic protein BAX, and increasing the anti-apoptotic protein BCL-2. Furthermore, the results showed that the GAS and GA combination could elevate the level of S1PR1 and inhibit the expression of ROCK2 and the phosphorylation of NF-κB, which are key targets involved in sphingolipid pathways. Conclusion Our study revealed that the combination of GAS and GA could suppress inflammation and apoptosis, which are highly correlated with sphingolipid signaling pathways, making it a potential candidate for the treatment of HTN.
Collapse
Affiliation(s)
- Shangtao Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Chenghao Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Shurui Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Siyu Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Baoshan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Shengbo Zhao
- Ningqiang Tianma Research Institution Limited Liability Company, Hanzhong, Shaanxi, People’s Republic of China
| | - Wei Zhang
- Ningqiang County Traditional Chinese Medicinal Industry Development Center, Hanzhong, Shaanxi, People’s Republic of China
| | - Zhirong Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
10
|
Mrugacz M, Zorena K, Pony-Uram M, Lendzioszek M, Pieńczykowska K, Bryl A. Interdependence of Nutrition, Physical Activity, and Myopia. Nutrients 2024; 16:3331. [PMID: 39408299 PMCID: PMC11478443 DOI: 10.3390/nu16193331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Myopia (also known as nearsightedness), a prevalent refractive error, occurs when parallel rays of light converge in front of the retina, resulting in blurry distance vision. Recently, there has been a marked rise in myopia among the global population. The absence of effective methods of controlling the progression of this visual defect prompts the search for new preventive and therapeutic options. The impact of diet and lifestyle on the progression of myopia is still not fully understood. Therefore, our aim was to examine how these factors might affect the advancement of myopia, based on the existing literature. Methods: This manuscript was prepared through an extensive literature review conducted from June 2022 to September 2024. We searched for pertinent research articles using reputable databases, including PubMed, Scopus, and Web of Science. We included all types of publications, with a special focus on the newest ones. Results: Despite far-reaching examination, the relationship between these factors and myopia control remains inconclusive with varying degrees of evidence supporting their roles. Conclusions: However, promoting a healthy lifestyle, particularly increasing physical activity and outdoor time, is essential. Additionally, emerging research suggests that maintaining a balanced diet is important due to the potential impact of certain nutrients on myopia development. Ophthalmologists should also guide parents on the alternative correction methods beyond single vision glasses, especially for rapidly progressing cases. With the rising prevalence of myopia in children, further research is necessary.
Collapse
Affiliation(s)
- Małgorzata Mrugacz
- Department of Ophthalmology and Eye Rehabilitation, Medical University of Bialystok, 15-089 Bialystok, Poland;
| | - Katarzyna Zorena
- Department of Immunobiology and Environmental Microbiology, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Magdalena Pony-Uram
- Department of Ophthalmology, Subcarpathian Hospital in Krosno, Korczynska 57, 38-400 Krosno, Poland;
| | - Maja Lendzioszek
- Department of Ophthalmology, Voivodship Hospital in Lomza, 18-400 Lomza, Poland;
| | | | - Anna Bryl
- Department of Ophthalmology and Eye Rehabilitation, Medical University of Bialystok, 15-089 Bialystok, Poland;
| |
Collapse
|
11
|
Hadidi M, Liñán-Atero R, Tarahi M, Christodoulou MC, Aghababaei F. The Potential Health Benefits of Gallic Acid: Therapeutic and Food Applications. Antioxidants (Basel) 2024; 13:1001. [PMID: 39199245 PMCID: PMC11352096 DOI: 10.3390/antiox13081001] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024] Open
Abstract
Gallic acid (GA), a phenolic acid found in fruits and vegetables, has been consumed by humans for centuries. Its extensive health benefits, such as antimicrobial, antioxidant, anticancer, anti-inflammatory, and antiviral properties, have been well-documented. GA's potent antioxidant capabilities enable it to neutralize free radicals, reduce oxidative stress, and protect cells from damage. Additionally, GA exerts anti-inflammatory effects by inhibiting inflammatory cytokines and enzymes, making it a potential therapeutic agent for inflammatory diseases. It also demonstrates anticancer properties by inhibiting cancer cell growth and promoting apoptosis. Furthermore, GA offers cardiovascular benefits, such as lowering blood pressure, decreasing cholesterol, and enhancing endothelial function, which may aid in the prevention and management of cardiovascular diseases. This review covers the chemical structure, sources, identification and quantification methods, and biological and therapeutic properties of GA, along with its applications in food. As research progresses, the future for GA appears promising, with potential uses in functional foods, pharmaceuticals, and nutraceuticals aimed at improving overall health and preventing disease. However, ongoing research and innovation are necessary to fully understand its functional benefits, address current challenges, and establish GA as a mainstay in therapeutic and nutritional interventions.
Collapse
Affiliation(s)
- Milad Hadidi
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Rafael Liñán-Atero
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, 13071 Ciudad Real, Spain;
| | - Mohammad Tarahi
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz 7144165186, Iran;
| | | | | |
Collapse
|
12
|
Cai Y, Zhao D, Pan Y, Chen B, Cao Y, Han S, Lian F, Zhang Y, Yan X. Gallic Acid Attenuates Sepsis-Induced Liver Injury through C/EBPβ-Dependent MAPK Signaling Pathway. Mol Nutr Food Res 2024; 68:e2400123. [PMID: 38809052 DOI: 10.1002/mnfr.202400123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/19/2024] [Indexed: 05/30/2024]
Abstract
SCOPE Liver injury is a major complication associated with sepsis. Together with others, the study has shown that gallic acid (GA) exerts anti-inflammatory and antioxidant effects in vivo. However, the role of GA in sepsis-mediated hepatic impairment and the underlying mechanisms remains to be elucidated. METHODS AND RESULTS C57BL/6J mice are pretreated with saline or GA and subjected to sham or cecal ligation and puncture (CLP). The pathological alterations are assessed by hematoxylin and eosin staining as well as immunohistochemical staining. RNA sequencing is employed to analyze hepatic transcriptome modifications. The study finds that GA supplementation significantly ameliorates CLP-induced mortality, liver dysfunction, and inflammation. RNA sequencing reveals that 1324 genes are markedly differentially regulated in livers of saline- or GA-treated sham or CLP mice. Gene ontology analysis demonstrates that the differentially expressed genes regulated by GA are predominantly correlated with the immune system process, oxidation-reduction process, and inflammatory response. Furthermore, mitogen-activated protein kinase (MAPK) signaling is localized in the center of the GA-mediated pathway network. Notably, activation of MAPK by C16-PAF significantly blocks GA-mediated protective effects on hepatic injury, inflammation, as well as CCAAT/enhancer-binding protein-β (C/EBPβ) dependent extracellular signal-regulated kinase 1/2 (ERK1/2) and nuclear factor-κB (NF-κB) signaling. CONCLUSION Therefore, this study indicates that GA may offer a promising therapeutic opportunity for sepsis-associated liver injury.
Collapse
Affiliation(s)
- Yuwei Cai
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Denghui Zhao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Yu Pan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Bingqi Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Yifei Cao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Shufen Han
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Fuzhi Lian
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| | - Yunlong Zhang
- Department of Cardiology, The First Affiliated Hospital (Yijishan Hospital) of Wannan Medical College, Wuhu, Anhui, 241001, China
| | - Xiao Yan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China, 311121
| |
Collapse
|
13
|
Sahu R, Rawal RK. Modulation of the c-JNK/p38-MAPK signaling pathway: Investigating the therapeutic potential of natural products in hypertension. PHYTOMEDICINE PLUS 2024; 4:100564. [DOI: 10.1016/j.phyplu.2024.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Ibrahim BMM, Elbaset MA, Abou Baker DH, Zikri EN, El Gengaihi S, Abdel Salam M. A pharmacological and toxicological biochemical study of cardiovascular regulatory effects of hibiscus, corn silk, marjoram, and chamomile. Heliyon 2024; 10:e22659. [PMID: 38226236 PMCID: PMC10788201 DOI: 10.1016/j.heliyon.2023.e22659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/17/2024] Open
Abstract
Hypertension is one of the most typical causes of morbidity and mortality. The present study investigated the possible antihypertensive cardiovascular effects of an herbal mixture extract of Hibiscus, Corn silk, Marjoram, and Chamomile. HPLC analysis of the water extract prepared from the aerial parts of four plants and their mixture was done to detect the most predominant compounds. A safety study was done prior to the efficacy study to determine the dose and ensure the extract's safety in female rats. Hypertension was induced in ovariectomized and non-ovariectomized rats by oral administration of 50 mg/kg of LName for 30 days; the hypertensive rats were classified into non-ovariectomized and ovariectomized untreated groups, treated groups with high and low doses of the mixture(150,300 mg/kg) given to ovariectomized and non-ovariectomized hypertensive groups and a standard group treated with angiotensin-converting enzyme inhibitor. The untreated group showed significant elevation of blood pressure, heart rate, cholesterol, triglycerides, malondialdehyde, cyclic adenosine monophosphate, angiotensin-converting enzyme, C-reactive protein, and significantly lowered reduced glutathione, high-density lipoprotein, and endothelial nitric oxide synthase. Treatment significantly counteracted the effects of L Name. The mixture provides a promising natural cardiovascular regulating supplement owing to its high contents of flavonoids.
Collapse
Affiliation(s)
- Bassant MM. Ibrahim
- Pharmacology Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki Giza, PO:12622, Egypt
| | - Marawan A. Elbaset
- Pharmacology Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki Giza, PO:12622, Egypt
| | - Doha H. Abou Baker
- Medicinal and Aromatic Plants Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Giza, Egypt, PO: 12622
| | - Emad N. Zikri
- Alternative and Complementary Medicine Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki Giza, PO:12622, Egypt
| | - Souad El Gengaihi
- Medicinal and Aromatic Plants Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Giza, Egypt, PO: 12622
| | - Mouchira Abdel Salam
- Alternative and Complementary Medicine Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki Giza, PO:12622, Egypt
| |
Collapse
|
15
|
Momeni Z, Danesh S, Ahmadpour M, Eshraghi R, Farkhondeh T, Pourhanifeh MH, Samarghandian S. Protective Roles and Therapeutic Effects of Gallic Acid in the Treatment of Cardiovascular Diseases: Current Trends and Future Directions. Curr Med Chem 2024; 31:3733-3751. [PMID: 37815180 DOI: 10.2174/0109298673259299230921150030] [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] [Received: 05/06/2023] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
Cardiovascular diseases (CVDs) are serious life-threatening illnesses and significant problematic issues for public health having a heavy economic burden on all society worldwide. The high incidence of these diseases as well as high mortality rates make them the leading causes of death and disability. Therefore, finding novel and more effective therapeutic methods is urgently required. Gallic acid, an herbal medicine with numerous biological properties, has been utilized in the treatment of various diseases for thousands of years. It has been demonstrated that gallic acid possesses pharmacological potential in regulating several molecular and cellular processes such as apoptosis and autophagy. Moreover, gallic acid has been investigated in the treatment of CVDs both in vivo and in vitro. Herein, we aimed to review the available evidence on the therapeutic application of gallic acid for CVDs including myocardial ischemia-reperfusion injury and infarction, drug-induced cardiotoxicity, hypertension, cardiac fibrosis, and heart failure, with a focus on underlying mechanisms.
Collapse
Affiliation(s)
- Zahra Momeni
- Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh Danesh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Ahmadpour
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Eshraghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeed Samarghandian
- University of Neyshabur Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
16
|
Shi Y, Liu C, Xiong S, Yang L, Yang C, Qiao W, Liu Y, Liu S, Liu J, Dong G. Ling-Gui-Qi-Hua formula alleviates left ventricular myocardial fibrosis in rats with heart failure with preserved ejection fraction by blocking the transforming growth factor-β1 /Smads signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116849. [PMID: 37385575 DOI: 10.1016/j.jep.2023.116849] [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: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ling-Qui-Qi-Hua (LGQH) decoction, composed of Poria cocos (Schw.) Wolf, Cinnamomum cassia (L.) J. Presl, Paeonia veitchii Lynch, and Atractylodes macrocephala Koidz., is a compound formula derived from Ling-Gui-Zhu-Gan decoction recorded in the Treatise on Febrile and Miscellaneous. It has shown cardioprotective effects on patients or rats with heart failure with preserved ejection fraction (HFpEF). Nevertheless, the active ingredients of LGQH and its anti-fibrotic mechanism remain unknown. AIM OF THE STUDY To determine the active ingredients in LGQH decoction and verify that LGQH decoction may inhibit left ventricular (LV) myocardial fibrosis in HFpEF rats by blocking the transforming growth factor-β1 (TGF-β1)/Smads signaling pathway from the perspective of animal experiments. MATERIALS AND METHODS First, liquid chromatography-mass spectrometry (LC-MS) technology was used to identify active components in the LGQH decoction. Secondly, a rat model of the metabolic syndrome-associated HFpEF phenotype was established and subsequently received LGQH intervention. The mRNA and protein expression of targets in the TGF-β1/Smads pathway were detected by quantitative real-time polymerase chain reaction and western blot analysis. Finally, molecular docking was conducted to examine the interactions between the active ingredients in the LGQH decoction and key proteins of the TGF-β1/Smads pathways. RESULTS According to LC-MS analysis, the LGQH decoction contained 13 active ingredients. In animal experiments, LGQH attenuated LV hypertrophy, enlargement, and diastolic function in HEpEF rats. Mechanically, LGQH not only down-regulated TGF-β1, Smad2, Smad3, Smad4, α-SMA, Coll I, and Coll III mRNA expressions and TGF-β1, Smad2, Smad3, P-Smad2/Smad3, Smad4, α-SMA, and Coll I protein expressions, but also up-regulated Smad7 mRNA and protein expressions, which ultimately led to myocardial fibrosis. Furthermore, molecular docking confirmed that 13 active ingredients in the LGQH decoction have excellent binding activities to the critical targets of the TGF-β1/Smads pathway. CONCLUSION LGQH is a modified herbal formulation with multiple active ingredients. It might alleviate LV remodeling and diastolic dysfunction and inhibit LV myocardial fibrosis by blocking TGF-β1/Smads pathways in HFpEF rats.
Collapse
Affiliation(s)
- Yujiao Shi
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Chunqiu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Shuang Xiong
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Ling Yang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Chenguang Yang
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Wenbo Qiao
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Yongcheng Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Siyu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Jiangang Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China.
| | - Guoju Dong
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China.
| |
Collapse
|
17
|
Akumwami S, Morishita A, Iradukunda A, Kobara H, Nishiyama A. Possible organ-protective effects of renal denervation: insights from basic studies. Hypertens Res 2023; 46:2661-2669. [PMID: 37532952 DOI: 10.1038/s41440-023-01393-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023]
Abstract
Inappropriate sympathetic nervous activation is the body's response to biological stress and is thought to be involved in the development of various lifestyle-related diseases through an elevation in blood pressure. Experimental studies have shown that surgical renal denervation decreases blood pressure in hypertensive animals. Recently, minimally invasive catheter-based renal denervation has been clinically developed, which results in a reduction in blood pressure in patients with resistant hypertension. Accumulating evidence in basic studies has shown that renal denervation exerts beneficial effects on cardiovascular disease and chronic kidney disease. Interestingly, recent studies have also indicated that renal denervation improves glucose tolerance and inflammatory changes. In this review article, we summarize the evidence from animal studies to provide comprehensive insight into the organ-protective effects of renal denervation beyond changes in blood pressure.
Collapse
Affiliation(s)
- Steeve Akumwami
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| |
Collapse
|
18
|
Elmileegy IMH, Waly HSA, Alghriany AAI, Abou Khalil NS, Mahmoud SMM, Negm EA. Gallic acid rescues uranyl acetate induced-hepatic dysfunction in rats by its antioxidant and cytoprotective potentials. BMC Complement Med Ther 2023; 23:423. [PMID: 37993853 PMCID: PMC10664358 DOI: 10.1186/s12906-023-04250-y] [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: 05/13/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND The liver was identified as a primary target organ for the chemo-radiological effects of uranyl acetate (UA). Although the anti-oxidant and anti-apoptotic properties of gallic acid (GA) make it a promising phytochemical to resist its hazards, there is no available data in this area of research. METHODS To address this issue, eighteen rats were randomly and equally divided into three groups. One group was received carboxymethyl cellulose (vehicle of GA) and kept as a control. The UA group was injected intraperitoneally with UA at a single dose of 5 mg/kg body weight. The third group (GA + UA group) was treated with GA orally at a dose of 100 mg/kg body weight for 14 days before UA exposure. UA was injected on the 15th day of the experiment in either the UA group or the GA + UA group. The biochemical, histological, and immunohistochemical findings in the GA + UA group were compared to both control and UA groups. RESULTS The results showed that UA exposure led to a range of adverse effects. These included elevated plasma levels of aspartate aminotransferase, lactate dehydrogenase, total protein, globulin, glucose, total cholesterol, triglycerides, low-density lipoprotein cholesterol, and very-low-density lipoprotein and decreased plasma levels of high-density lipoprotein cholesterol. The exposure also disrupted the redox balance, evident through decreased plasma total antioxidant capacity and hepatic nitric oxide, superoxide dismutase, reduced glutathione, glutathione-S-transferase, glutathione reductase, and glutathione peroxidase and increased hepatic oxidized glutathione and malondialdehyde. Plasma levels of albumin and alanine aminotransferase did not significantly change in all groups. Histopathological analysis revealed damage to liver tissue, characterized by deteriorations in tissue structure, excessive collagen accumulation, and depletion of glycogen. Furthermore, UA exposure up-regulated the immuno-expression of cleaved caspase-3 and down-regulated the immuno-expression of nuclear factor-erythroid-2-related factor 2 in hepatic tissues, indicating an induction of apoptosis and oxidative stress response. However, the pre-treatment with GA proved to be effective in mitigating these negative effects induced by UA exposure, except for the disturbances in the lipid profile. CONCLUSIONS The study suggests that GA has the potential to act as a protective agent against the adverse effects of UA exposure on the liver. Its ability to restore redox balance and inhibit apoptosis makes it a promising candidate for countering the harmful effects of chemo-radiological agents such as UA.
Collapse
Affiliation(s)
- Ibtisam M H Elmileegy
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt
| | - Hanan S A Waly
- Laboratory of Physiology, Department of Zoology and Entomology, Faculty of Science, Assiut University, Assiut, Egypt
| | | | - Nasser S Abou Khalil
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt.
- Department of Basic Medical Sciences, Faculty of Physical Therapy, Merit University, Sohag, Egypt.
| | - Sara M M Mahmoud
- Department of Physiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Eman A Negm
- Department of Physiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
19
|
Bai L, Han X, Kee HJ, He X, Kim SH, Jeon MJ, Zhou H, Jeong SM, Kee SJ, Jeong MH. Protocatechuic acid prevents isoproterenol-induced heart failure in mice by downregulating kynurenine-3-monooxygenase. J Cell Mol Med 2023; 27:2290-2307. [PMID: 37482908 PMCID: PMC10424289 DOI: 10.1111/jcmm.17869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/12/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023] Open
Abstract
Protocatechuic acid (3,4-dihydroxybenzoic acid) prevents oxidative stress, inflammation and cardiac hypertrophy. This study aimed to investigate the therapeutic effects of protocatechuic acid in an isoproterenol-induced heart failure mouse model and to identify the underlying mechanisms. To establish the heart failure model, C57BL/6NTac mice were given high-dose isoproterenol (80 mg/kg body weight) for 14 days. Echocardiography revealed that protocatechuic acid reversed the isoproterenol-induced downregulation of fractional shortening and ejection fraction. Protocatechuic acid attenuated cardiac hypertrophy as evidenced by the decreased heart-weight-to-body-weight ratio and the expression of Nppb. RNA sequencing analysis identified kynurenine-3-monooxygenase (Kmo) as a potential target of protocatechuic acid. Protocatechuic acid treatment or transfection with short-interfering RNA against Kmo ameliorated transforming growth factor β1-induced upregulation of Kmo, Col1a1, Col1a2 and Fn1 in vivo or in neonatal rat cardiac fibroblasts. Kmo knockdown attenuated the isoproterenol-induced increase in cardiomyocyte size, as well as Nppb and Col1a1 expression in H9c2 cells or primary neonatal rat cardiomyocytes. Moreover, protocatechuic acid attenuated Kmo overexpression-induced increases in Nppb mRNA levels. Protocatechuic acid or Kmo knockdown decreased isoproterenol-induced ROS generation in vivo and in vitro. Thus, protocatechuic acid prevents heart failure by downregulating Kmo. Therefore, protocatechuic acid and Kmo constitute a potential novel therapeutic agent and target, respectively, against heart failure.
Collapse
Affiliation(s)
- Liyan Bai
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
- Emergency Critical Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiongyi Han
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
- Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, People's Republic of China
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Xiaonan He
- Emergency Critical Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Seong Hoon Kim
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Mi Jin Jeon
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hongyan Zhou
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seong Min Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
20
|
Su HX, Li PB, Shi KN, Gao J, Zhang HJ, Li HH. The immunoproteasome subunit β2i ameliorates myocardial ischemia/reperfusion injury by regulating Parkin-Mfn1/2-mediated mitochondrial fusion. Cell Mol Life Sci 2023; 80:231. [PMID: 37501008 PMCID: PMC11071955 DOI: 10.1007/s00018-023-04867-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/02/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023]
Abstract
Mitochondrial dynamics are critical for maintaining mitochondrial morphology and function during cardiac ischemia and reperfusion (I/R). The immunoproteasome complex is an inducible isoform of the proteasome that plays a key role in modulating inflammation and some cardiovascular diseases, but the importance of immunoproteasome catalytic subunit β2i (also known as LMP10 or MECL1) in regulating mitochondrial dynamics and cardiac I/R injury is largely unknown. Here, using β2i-knockout (KO) mice and rAAV9-β2i-injected mice, we discovered that β2i expression and its trypsin-like activity were significantly attenuated in the mouse I/R myocardium and in patients with myocardial infarction (MI). Moreover, β2i-KO mice exhibited greatly enhanced I/R-mediated cardiac dysfunction, infarct size, myocyte apoptosis and oxidative stress accompanied by excessive mitochondrial fission due to Mfn1/2 and Drp1 imbalance. Conversely, cardiac overexpression of β2i in mice injected with recombinant adeno-associated virus 9 (rAAV9)-β2i ameliorated cardiac I/R injury. Mechanistically, I/R injury reduced β2i expression and activity, which increased the expression of the E3 ligase Parkin protein and promoted the degradation of mitofusin 1/2 (Mfn1/2), leading to excessive mitochondrial fission. In conclusion, our data suggest for the first time that β2i exerts a protective role against cardiac I/R injury and that increasing β2i expression may be a new therapeutic option for cardiac ischemic disease in clinical practice. Graphical abstract showing how the immunoproteasome subunit β2i ameliorates myocardial I/R injury by regulating Parkin-Mfn1/2-mediated mitochondrial fusion.
Collapse
Affiliation(s)
- Hui-Xiang Su
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing, 100020, China
| | - Pang-Bo Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing, 100020, China
| | - Kai-Na Shi
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing, 100020, China
| | - Jing Gao
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing, 100020, China
| | - Hong-Jia Zhang
- Department of Cardiac Surgery, Beijing An-Zhen Hospital, Capital Medical University, No. 2 An-Zhen Road, Chaoyang District, Beijing, 100029, China.
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing, 100020, China.
| |
Collapse
|
21
|
Festa J, Hussain A, Al-Hareth Z, Singh H, Da Boit M. Anthocyanins and Vascular Health: A Matter of Metabolites. Foods 2023; 12:foods12091796. [PMID: 37174334 PMCID: PMC10178014 DOI: 10.3390/foods12091796] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Anthocyanins are a subgroup of flavonoid polyphenols previously investigated for improving cardiovascular health and preventing the development of endothelial dysfunction. However, their poor bioavailability raises the question of whether the observed biological activity is due to their metabolites. Phenolic metabolites can reach higher plasma concentrations and can persist in the circulation for periods much longer than their original anthocyanin form; therefore, the biological activity and health promoting effects of anthocyanins may differ from their metabolites. To address this, recent studies have facilitated different cell models, in vivo studies and explored physiologically relevant concentrations to better understand their mechanisms of action. The criteria were chosen based on previous reports demonstrating that anthocyanins can improve endothelial function via modulation of the Akt-endothelial nitric oxide synthase pathway and transcription factors Nrf2 and NF-κB, which made it critical to assess the phenolic metabolites' modes of action via these pathways. This review demonstrates how phenolic metabolites differ in bioactivity from their precursor anthocyanin, demonstrating improved endothelial function in response to inflammatory mediators at concentrations that are tolerated in vivo. The review highlights the crucial need for further studies to focus on improving the bioavailability of metabolites in isolation and explore the effect of metabolites in mixtures.
Collapse
Affiliation(s)
- Joseph Festa
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Aamir Hussain
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Zakia Al-Hareth
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
- Pandemic Sciences Institute, Old Road Campus, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
| | - Harprit Singh
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Mariasole Da Boit
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| |
Collapse
|
22
|
Yu M, Kim HJ, Heo H, Kim M, Jeon Y, Lee H, Lee J. Comparison of the Antihypertensive Activity of Phenolic Acids. Molecules 2022; 27:molecules27196185. [PMID: 36234721 PMCID: PMC9571617 DOI: 10.3390/molecules27196185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022] Open
Abstract
Phenolic acids, found in cereals, legumes, vegetables, and fruits, have various biological functions. We aimed to compare the antihypertensive potential of different phenolic acids by evaluating their ACE inhibitory activity and cytoprotective capacity in EA.hy 926 endothelial cells. In addition, we explored the mechanism underlying the antihypertensive activity of sinapic acid. Of all the phenolic acids studied, sinapic acid, caffeic acid, coumaric acid, and ferulic acid significantly inhibited ACE activity. Moreover, gallic acid, sinapic acid, and ferulic acid significantly enhanced intracellular NO production. Based on the results of GSH depletion, ROS production, and MDA level analyses, sinapic acid was selected to study the mechanism underlying the antihypertensive effect. Sinapic acid decreases endothelial dysfunction by enhancing the expression of antioxidant-related proteins. Sinapic acid increased phosphorylation of eNOS and Akt in a dose-dependent manner. These findings indicate the potential of sinapic acid as a treatment for hypertension.
Collapse
Affiliation(s)
- Myeongnam Yu
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Hyun Joo Kim
- Department of Central Area Crop Science, National Institute of Crop Science, Rural Development Administration, Suwon 16613, Korea
| | - Huijin Heo
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Minjun Kim
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Yesol Jeon
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Hana Lee
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
- Correspondence: (H.L.); (J.L.)
| | - Junsoo Lee
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
- Correspondence: (H.L.); (J.L.)
| |
Collapse
|
23
|
Yan X, Zhang YL, Han X, Li PB, Guo SB, Li HH. Time Series Transcriptomic Analysis by RNA Sequencing Reveals a Key Role of PI3K in Sepsis-Induced Myocardial Injury in Mice. Front Physiol 2022; 13:903164. [PMID: 35721566 PMCID: PMC9198581 DOI: 10.3389/fphys.2022.903164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Septic cardiomyopathy is the main complication and cause of death of severe sepsis with limited therapeutic strategy. However, the molecular mechanism of sepsis-induced cardiac injury remains unclear. The present study was designed to investigate differentially expressed genes (DEGs) involved in the pathogenesis of septic cardiomyopathy induced by cecal ligation and puncture (CLP) in mice. Male C57BL/6J mice (8-10 weeks old) were subjected to CLP with 21-gauge needles for 24, 48, and 72 h. Myocardial function was assessed by echocardiography. The pathological changes of the heart were evaluated by hematoxylin and eosin as well as immunohistochemical staining. Time series RNA sequencing was utilized to investigate the gene expression profiles. CLP surgery resulted in a significant decrease of animal survival rate and left ventricle contractile function, and an increase in cardiac dilation and infiltration of proinflammatory cells including Mac-2+ macrophages in a time-dependent manner. RNA sequencing identified 5,607 DEGs in septic myocardium at 24, 48, and 72 h after CLP operation. Moreover, gene ontology analysis revealed that these DEGs were mainly associated with the biological processes, including cell adhesion, immune system process, inflammatory response, and positive regulation of cell migration. KEGG pathway enrichment analysis indicated that Staphylococcus aureus infection, osteoclast differentiation, leishmaniasis, and ECM-receptor interaction were significantly altered in septic hearts. Notably, Pik3r1 and Pik3r5 were localized in the center of the gene co-expression network, and were markedly upregulated in CLP-induced septic myocardium. Further, blocking PI3Kγ by the specific inhibitor CZC24832 significantly protected against sepsis-induced cardiac impairment. The present study uncovers the gene expression signatures of CLP-induced myocardial injury and sheds light on the role of Pik3r5 in septic cardiomyopathy.
Collapse
Affiliation(s)
- Xiao Yan
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China.,School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yun-Long Zhang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Xiao Han
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Pang-Bo Li
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Shu-Bin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Hui-Hua Li
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| |
Collapse
|
24
|
Han X, Bai L, Kee HJ, Jeong MH. Syringic acid mitigates isoproterenol-induced cardiac hypertrophy and fibrosis by downregulating Ereg. J Cell Mol Med 2022; 26:4076-4086. [PMID: 35719043 PMCID: PMC9279583 DOI: 10.1111/jcmm.17449] [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: 03/24/2022] [Revised: 04/22/2022] [Accepted: 05/11/2022] [Indexed: 12/01/2022] Open
Abstract
Gallic acid has been reported to mitigate cardiac hypertrophy, fibrosis and arterial hypertension. The effects of syringic acid, a derivative of gallic acid, on cardiac hypertrophy and fibrosis have not been previously investigated. This study aimed to examine the effects of syringic acid on isoproterenol‐treated mice and cells. Syringic acid mitigated the isoproterenol‐induced upregulation of heart weight to bodyweight ratio, pathological cardiac remodelling and fibrosis in mice. Picrosirius red staining, quantitative real‐time polymerase chain reaction (qRT‐PCR) and Western blotting analyses revealed that syringic acid markedly downregulated collagen accumulation and fibrosis‐related factors, including Fn1. The results of RNA sequencing analysis of Ereg expression were verified using qRT‐PCR. Syringic acid or transfection with si‐Ereg mitigated the isoproterenol‐induced upregulation of Ereg, Myc and Ngfr. Ereg knockdown mitigated the isoproterenol‐induced upregulation of Nppb and Fn1 and enhancement of cell size. Mechanistically, syringic acid alleviated cardiac hypertrophy and fibrosis by downregulating Ereg. These results suggest that syringic acid is a potential therapeutic agent for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Xiongyi Han
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Liyan Bai
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea.,Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
25
|
Zinc-dependent histone deacetylases: Potential therapeutic targets for arterial hypertension. Biochem Pharmacol 2022; 202:115111. [DOI: 10.1016/j.bcp.2022.115111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022]
|
26
|
Angiotensin II Induces Cardiac Edema and Hypertrophic Remodeling through Lymphatic-Dependent Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5044046. [PMID: 35222798 PMCID: PMC8881141 DOI: 10.1155/2022/5044046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/17/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022]
Abstract
Cardiac lymphatic vessel growth (lymphangiogenesis) and integrity play an essential role in maintaining tissue fluid balance. Inhibition of lymphatic lymphangiogenesis is involved in cardiac edema and cardiac remodeling after ischemic injury or pressure overload. However, whether lymphatic vessel integrity is disrupted during angiotensin II- (Ang II-) induced cardiac remodeling remains to be investigated. In this study, cardiac remodeling models were established by Ang II (1000 ng/kg/min) in VEGFR-3 knockdown (Lyve-1Cre VEGFR-3f/−) and wild-type (VEGFR-3f/f) littermates. Our results indicated that Ang II infusion not only induced cardiac lymphangiogenesis and upregulation of VEGF-C and VEGFR-3 expression in the time-dependent manner but also enhanced proteasome activity, MKP5 and VE-cadherin degradation, p38 MAPK activation, and lymphatic vessel hyperpermeability. Moreover, VEGFR-3 knockdown significantly inhibited cardiac lymphangiogenesis in mice, resulting in exacerbation of tissue edema, hypertrophy, fibrosis superoxide production, inflammation, and heart failure (HF). Conversely, administration of epoxomicin (a selective proteasome inhibitor) markedly mitigated Ang II-induced cardiac edema, remodeling, and dysfunction; upregulated MKP5 and VE-cadherin expression; inactivated p38 MAPK; and reduced lymphatic vessel hyperpermeability in WT mice, indicating that inhibition of proteasome activity is required to maintain lymphatic endothelial cell (LEC) integrity. Our results show that both cardiac lymphangiogenesis and lymphatic barrier hyperpermeability are implicated in Ang II-induced adaptive hypertrophic remodeling and dysfunction. Proteasome-mediated hyperpermeability of LEC junctions plays a predominant role in the development of cardiac remodeling. Selective stimulation of lymphangiogenesis or inhibition of proteasome activity may be a potential therapeutic option for treating hypertension-induced cardiac remodeling.
Collapse
|
27
|
Peroxiredoxin-5 Knockdown Accelerates Pressure Overload-Induced Cardiac Hypertrophy in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5067544. [PMID: 35132351 PMCID: PMC8817848 DOI: 10.1155/2022/5067544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022]
Abstract
A recent study showed that peroxiredoxins (Prxs) play an important role in the development of pathological cardiac hypertrophy. However, the involvement of Prx5 in cardiac hypertrophy remains unclear. Therefore, this study is aimed at investigating the role and mechanisms of Prx5 in pathological cardiac hypertrophy and dysfunction. Transverse aortic constriction (TAC) surgery was performed to establish a pressure overload-induced cardiac hypertrophy model. In this study, we found that Prx5 expression was upregulated in hypertrophic hearts and cardiomyocytes. In addition, Prx5 knockdown accelerated pressure overload-induced cardiac hypertrophy and dysfunction in mice by activating oxidative stress and cardiomyocyte apoptosis. Importantly, heart deterioration caused by Prx5 knockdown was related to mitogen-activated protein kinase (MAPK) pathway activation. These findings suggest that Prx5 could be a novel target for treating cardiac hypertrophy and heart failure.
Collapse
|
28
|
Ashrafizadeh M, Zarrabi A, Mirzaei S, Hashemi F, Samarghandian S, Zabolian A, Hushmandi K, Ang HL, Sethi G, Kumar AP, Ahn KS, Nabavi N, Khan H, Makvandi P, Varma RS. Gallic acid for cancer therapy: Molecular mechanisms and boosting efficacy by nanoscopical delivery. Food Chem Toxicol 2021; 157:112576. [PMID: 34571052 DOI: 10.1016/j.fct.2021.112576] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/23/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Cancer is the second leading cause of death worldwide. Majority of recent research efforts in the field aim to address why cancer resistance to therapy develops and how to overcome or prevent it. In line with this, novel anti-cancer compounds are desperately needed for chemoresistant cancer cells. Phytochemicals, in view of their pharmacological activities and capacity to target various molecular pathways, are of great interest in the development of therapeutics against cancer. Plant-derived-natural products have poor bioavailability which restricts their anti-tumor activity. Gallic acid (GA) is a phenolic acid exclusively found in natural sources such as gallnut, sumac, tea leaves, and oak bark. In this review, we report on the most recent research related to anti-tumor activities of GA in various cancers with a focus on its underlying molecular mechanisms and cellular pathwaysthat that lead to apoptosis and migration of cancer cells. GA down-regulates the expression of molecular pathways involved in cancer progression such as PI3K/Akt. The co-administration of GA with chemotherapeutic agents shows improvements in suppressing cancer malignancy. Various nano-vehicles such as organic- and inorganic nano-materials have been developed for targeted delivery of GA at the tumor site. Here, we suggest that nano-vehicles improve GA bioavailability and its ability for tumor suppression.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey; Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Farid Hashemi
- Phd student of pharmacology, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| | - Pooyan Makvandi
- Centre for Materials Interfaces, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy.
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
29
|
Potential Role of Melatonin as an Adjuvant for Atherosclerotic Carotid Arterial Stenosis. Molecules 2021; 26:molecules26040811. [PMID: 33557283 PMCID: PMC7914857 DOI: 10.3390/molecules26040811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/23/2022] Open
Abstract
Carotid artery stenosis (CAS) is an atherosclerotic disease characterized by a narrowing of the artery lumen and a high risk of ischemic stroke. Risk factors of atherosclerosis, including smoking, hypertension, hyperglycemia, hyperlipidemia, aging, and disrupted circadian rhythm, may potentiate atherosclerosis in the carotid artery and further reduce the arterial lumen. Ischemic stroke due to severe CAS and cerebral ischemic/reperfusion (I/R) injury after the revascularization of CAS also adversely affect clinical outcomes. Melatonin is a pluripotent agent with potent anti-inflammatory, anti-oxidative, and neuroprotective properties. Although there is a shortage of direct clinical evidence demonstrating the benefits of melatonin in CAS patients, previous studies have shown that melatonin may be beneficial for patients with CAS in terms of reducing endothelial damage, stabilizing arterial plaque, mitigating the harm from CAS-related ischemic stroke and cerebral I/R injury, and alleviating the adverse effects of the related risk factors. Additional pre-clinical and clinical are required to confirm this speculation.
Collapse
|
30
|
Najjar RS, Turner CG, Wong BJ, Feresin RG. Berry-Derived Polyphenols in Cardiovascular Pathologies: Mechanisms of Disease and the Role of Diet and Sex. Nutrients 2021; 13:nu13020387. [PMID: 33513742 PMCID: PMC7911141 DOI: 10.3390/nu13020387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) prevalence, pathogenesis, and manifestation is differentially influenced by biological sex. Berry polyphenols target several signaling pathways pertinent to CVD development, including inflammation, oxidative stress, and cardiac and vascular remodeling, and there are innate differences in these pathways that also vary by sex. There is limited research systematically investigating sex differences in berry polyphenol effects on these pathways, but there are fundamental findings at this time that suggest a sex-specific effect. This review will detail mechanisms within these pathological pathways, how they differ by sex, and how they may be individually targeted by berry polyphenols in a sex-specific manner. Because of the substantial polyphenolic profile of berries, berry consumption represents a promising interventional tool in the treatment and prevention of CVD in both sexes, but the mechanisms in which they function within each sex may vary.
Collapse
Affiliation(s)
- Rami S. Najjar
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA;
| | - Casey G. Turner
- Department of Kinesiology and Health, Georgia State University, Atlanta, GA 30302, USA; (C.G.T.); (B.J.W.)
| | - Brett J. Wong
- Department of Kinesiology and Health, Georgia State University, Atlanta, GA 30302, USA; (C.G.T.); (B.J.W.)
| | - Rafaela G. Feresin
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA;
- Correspondence:
| |
Collapse
|
31
|
Han D, Zhang QY, Zhang YL, Han X, Guo SB, Teng F, Yan X, Li HH. Gallic Acid Ameliorates Angiotensin II-Induced Atrial Fibrillation by Inhibiting Immunoproteasome- Mediated PTEN Degradation in Mice. Front Cell Dev Biol 2020; 8:594683. [PMID: 33251220 PMCID: PMC7673442 DOI: 10.3389/fcell.2020.594683] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/13/2020] [Indexed: 11/13/2022] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia and is a major cause of stroke and heart failure. We and others have found that gallic acid (GA) plays a beneficial role in cardiac hypertrophic remodeling and hypertension. However, the effect of GA on angiotensin II (Ang II)-induced AF and atrial remodeling as well as the underlying mechanisms remain unknown. AF was induced in mice by Ang II infusion (2000 ng/kg/min) for 3 weeks. Blood pressure was measured using the tail-cuff method. Atrial volume was evaluated by echocardiography. Atrial remodeling was studied using hematoxylin and eosin, Masson's trichrome, and immunohistochemical staining. Atrial oxidative stress was assessed by dihydroethidium staining. The gene expression of fibrotic and inflammatory markers and protein levels of signaling mediators were measured by quantitative real-time PCR and western blot analysis. In mice, GA administration significantly attenuated Ang II-induced elevation of blood pressure, AF incidence and duration, atrial dilation, fibrosis, inflammation, and oxidative stress compared with the vehicle control. Furthermore, GA downregulated Ang II-induced activity and expression of immunoproteasome subunits (β2i and β5i), which reduced PTEN degradation and led to the inactivation of AKT1 and downstream signaling mediators. Importantly, blocking PTEN activity by VO-Ohpic markedly reversed the GA-mediated protective effects on Ang II-induced AF and atrial remodeling. Therefore, our results provide novel evidence that GA exerts a cardioprotective role by inhibiting immunoproteasome activity, which attenuates PTEN degradation and activation of downstream signaling, and may represent a promising candidate for treating hypertensive AF.
Collapse
Affiliation(s)
- Dan Han
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi-Yu Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Zhang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Xiao Han
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Shu-Bin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Fei Teng
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Xiao Yan
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| |
Collapse
|