1
|
Yuan C, Wang Y, Zhang L, Wang D. Procatechuic acid and protocatechuic aldehyde increase survival of Caenorhabditis elegans after fungal infection and inhibit fungal virulence. Front Pharmacol 2024; 15:1396733. [PMID: 38841375 PMCID: PMC11150623 DOI: 10.3389/fphar.2024.1396733] [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: 03/06/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Protocatechuic acid (PCA) and protocatechuic aldehyde (PAL) are important phenolic compounds in plants. We here investigated their possible beneficial effect against fungal infection and the underlying mechanism. The model animal of Caenorhabditis elegans was used as host, and Candida albicans was used as fungal pathogen. The nematodes were first infected with C. albicans, and the PCA and PAL treatment were then performed. Post-treatment with 10-100 μM PCA and PAL suppressed toxicity of C. albicans infection in reducing lifespan. Accompanied with this beneficial effect, treatment with 10-100 μM PCA and PAL inhibited C. albicans accumulation in intestinal lumen. In addition, treatment with 10-100 μM PCA and PAL suppressed the increase in expressions of antimicrobial genes caused by C. albicans infection. The beneficial effect of PCA and PAL against C. albicans infection depended on p38 MAPK and insulin signals. Moreover, although treatment with 10-100 μM PCA and PAL could not exhibit noticeable antifungal activity, PCA and PAL treatment obviously suppressed biofilm formation, inhibited hyphal growth, and reduced expressions of virulence genes (ALS3, CaVps34, Vma7, Vac1, and/or HWP1) related to biofilm formation and hyphal growth in C. albicans. Therefore, our data demonstrated the potential of PCA and PAL post-treatment against fungal infection and fungal virulence.
Collapse
Affiliation(s)
- Chunyan Yuan
- Department of Gynaecology and Obstetrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yuxing Wang
- Deaprtment of Biochemistry and Molecrla Biology, School of Medicine, Southeast University, Nanjing, China
| | - Le Zhang
- Deaprtment of Biochemistry and Molecrla Biology, School of Medicine, Southeast University, Nanjing, China
| | - Dayong Wang
- Deaprtment of Biochemistry and Molecrla Biology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
2
|
Chen J, Zhao R, Wang Y, Xiao H, Lin W, Diao M, He S, Mei P, Liao Y. G protein-coupled estrogen receptor activates PI3K/AKT/mTOR signaling to suppress ferroptosis via SREBP1/SCD1-mediated lipogenesis. Mol Med 2024; 30:28. [PMID: 38383297 PMCID: PMC10880371 DOI: 10.1186/s10020-023-00763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/25/2023] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related death worldwide. The sex differences in the occurrence and fatality rates of non-small cell lung cancer (NSCLC), along with its association with estrogen dependence, suggest that estrogen receptors (ERs) contribute to the development of NSCLC. However, the influence of G protein-coupled estrogen receptor (GPER1) on NSCLC remains to be determined. Escape from ferroptosis is one of the hallmarks of tumor discovered in recent years. In this context, the present study evaluated whether GPER1 promotes NSCLC progression by preventing ferroptosis, and the underlying mechanism through which GPER1 protects against ferroptosis was also explored. METHODS The effects of GPER1 on the cytotoxicity of H2O2, the ferroptosis inducer RSL3, and Erastin were assessed using the CCK8 assay and plate cloning. Lipid peroxidation levels were measured based on the levels of MDA and BODIPY™581/591C11. GPER1 overexpression and knockdown were performed and G1 was used, and the expression of SCD1 and PI3K/AKT/mTOR signaling factors was measured. Immunofluorescence analysis and immunohistochemistry were performed on paired specimens to measure the correlation between the expression of GPER1 and SCD1 in NSCLC tissues. The effect of GPER1 on the cytotoxicity of cisplatin was measured in vitro using the CCK8 assay and in vivo using xenograft tumor models. RESULTS GPER1 and G1 alleviated the cytotoxicity of H2O2, reduced sensitivity to RSL3, and impaired lipid peroxidation in NSCLC tissues. In addition, GPER1 and G1 promoted the protein and mRNA expression of SCD1 and the activation of PI3K/AKT/mTOR signaling. GPER1 and SCD1 expression were elevated and positively correlated in NSCLC tissues, and high GPER1 expression predicted a poor prognosis. GPER1 knockdown enhanced the antitumor activity of cisplatin in vitro and in vivo. CONCLUSION GPER1 prevents ferroptosis in NSCLC by promoting the activation of PI3K/AKT/mTOR signaling, thereby inducing SCD1 expression. Therefore, treatments targeting GPER1 combined with cisplatin would exhibit better antitumor effects.
Collapse
Affiliation(s)
- Jiaping Chen
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Cardiothoracic Surgery, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, China
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Han Xiao
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Lin
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mingxin Diao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shiwen He
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Lu M, Pan J, Hu Y, Ding L, Li Y, Cui X, Zhang M, Zhang Z, Li C. Advances in the study of vascular related protective effect of garlic (Allium sativum) extract and compounds. J Nutr Biochem 2024; 124:109531. [PMID: 37984733 DOI: 10.1016/j.jnutbio.2023.109531] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 11/22/2023]
Abstract
Garlic (Allium sativum) is a functional food containing multiple bioactive compounds that find widespread applications in culinary and medicinal practices. It consists of multiple chemical components, including allicin and alliin. This article offers a comprehensive review of the protective effects of garlic extracts and their active constituents on the vascular system. In vitro and in vivo experiments have shown that garlic extracts and their active ingredients possess various bioactive properties. These substances demonstrate beneficial effects on blood vessels by demonstrating anti-inflammatory and antioxidant activities, inhibiting lipid accumulation and migration, preventing lipid peroxidation, promoting angiogenesis, reducing platelet aggregation, enhancing endothelial function, and inhibiting endothelial cell apoptosis. In clinical studies, garlic and its extracts have demonstrated their efficacy in managing vascular system diseases, including atherosclerosis, diabetes, and high cholesterol levels. In summary, these studies highlight the potential therapeutic roles and underlying mechanisms of garlic and its constituents in managing conditions like diabetes, atherosclerosis, ischemic diseases, and other vascular disorders.
Collapse
Affiliation(s)
- Mengkai Lu
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinyuan Pan
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liang Ding
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
4
|
Membrane damage mechanism of protocatechualdehyde against Micrococcus luteus and its effect on pork quality characteristics. Sci Rep 2022; 12:18856. [PMID: 36344587 PMCID: PMC9640607 DOI: 10.1038/s41598-022-23309-3] [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: 06/01/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
This study investigated the mechanism of membrane damage by protocatechualdehyde (PCA) against Micrococcus luteus and assessed effects of PCA on the sensory and physicochemical properties of pork. The mechanism of PCA inhibition on M. luteus was studied by determining the minimum inhibitory concentration (MIC) based on membrane potential, intracellular ATP concentration, intracellular pH, confocal laser scanning microscopy (CLSM), and field emission gun scanning electron microscopy (FEG-SEM). The results showed that the MIC of PCA against M. luteus was 1.25 mg/mL. Hyperpolarization of the bacterial cell membrane, a decrease in the intracellular ATP concentration, and intracellular pH indicated that PCA damaged the cell membrane of M. luteus. FEG-SEM observation revealed that PCA could cause surface collapse, cell membrane rupture, and content outflow of M. luteus. Additionally, PCA was found to inhibit increases in the total number of colonies, the thiobarbituric acid reactive substances (TBARS) value growth rate, and moisture mobility in raw pork. Additionally, it improved the color and texture of raw pork, all of which effectively prolonged its shelf life. This study will encourage the application of PCA as a natural antibacterial agent in the food industry.
Collapse
|
5
|
Liu H, Zhu L, Chen L, Li L. Therapeutic potential of traditional Chinese medicine in atherosclerosis: A review. Phytother Res 2022; 36:4080-4100. [PMID: 36029188 DOI: 10.1002/ptr.7590] [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: 04/05/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 11/12/2022]
Abstract
Atherosclerosis is the onset of endothelial cell damage and is characterized by abnormal accumulation of fibrinogen and lipid in large and middle arteries. Recent researches indicate that traditional Chinese medicine including Notoginseng Radix et Rhizoma, Astragali Radix, Salviae Miltiorrhizae Radix et Rhizoma, Ginseng Radix et Rhizoma, Fructus Crataegi, Glycyrrhizae Radix et Rhizoma, Polygoni Multiflori Radix, Fructus Lycii, and Coptidis Rhizoma have therapeutic effects on atherosclerosis. Furthermore, the pharmacological roles of these kinds of traditional Chinese medicine in atherosclerosis refer to endothelial function influences, cell proliferation and migration, platelet aggregation, thrombus formation, oxidative stress, inflammation, angiogenesis, apoptosis, autophagy, lipid metabolism, and the gut microbiome. Traditional Chinese medicine may serve as potential and effective anti-atherosclerosis drugs. However, a critical study has shown that Notoginseng Radix et Rhizoma may also have toxic effects including pustules, fever, and elevate circulating neutrophil count. Further high-quality studies are still required to determine the clinical safety and efficacy of traditional Chinese medicine and its active ingredients.
Collapse
Affiliation(s)
- Huimei Liu
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Zhu
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
6
|
G-Protein-Coupled Estrogen Receptor Expression in Rat Uterine Artery Is Increased by Pregnancy and Induces Dilation in a Ca2+ and ERK1/2 Dependent Manner. Int J Mol Sci 2022; 23:ijms23115996. [PMID: 35682675 PMCID: PMC9180712 DOI: 10.3390/ijms23115996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Increasing levels of estrogens across gestation are partly responsible for the physiological adaptations of the maternal vasculature to pregnancy. The G protein-coupled estrogen receptor (GPER) mediates acute vasorelaxing effects in the uterine vasculature, which may contribute to the regulation of uteroplacental blood flow. The aim of this study was to investigate whether GPER expression and vasorelaxation may occur following pregnancy. Elucidation of the functional signalling involved was also investigated. Radial uterine and third-order mesenteric arteries were isolated from non-pregnant (NP) and pregnant rats (P). GPER mRNA levels were determined and—concentration–response curve to the GPER-specific agonist, G1 (10−10–10−6 M), was assessed in arteries pre-constricted with phenylephrine. In uterine arteries, GPER mRNA expression was significantly increased and vasorelaxation to G1 was significantly enhanced in P compared with NP rats. Meanwhile, in mesenteric arteries, there was a similar order of magnitude in NP and P rats. Inhibition of L-type calcium channels and extracellular signal-regulated kinases 1/2 significantly reduced vasorelaxation triggered by G1 in uterine arteries. Increased GPER expression and GPER-mediated vasorelaxation are associated with the advancement of gestation in uterine arteries. The modulation of GPER is exclusive to uterine arteries, thus suggesting a physiological contribution of GPER toward the regulation of uteroplacental blood flow during pregnancy.
Collapse
|
7
|
Soheili M, Alinaghipour A, Salami M. Good bacteria, oxidative stress and neurological disorders: Possible therapeutical considerations. Life Sci 2022; 301:120605. [DOI: 10.1016/j.lfs.2022.120605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022]
|
8
|
The combination of four main components in Xuebijing injection improved the preventive effects of Cyclosporin A in acute graft-versus-host disease mice by protecting intestinal microenvironment. Pharmacotherapy 2022; 148:112675. [DOI: 10.1016/j.biopha.2022.112675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/28/2022]
|
9
|
Zhang S, Gai Z, Gui T, Chen J, Chen Q, Li Y. Antioxidant Effects of Protocatechuic Acid and Protocatechuic Aldehyde: Old Wine in a New Bottle. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6139308. [PMID: 34790246 PMCID: PMC8592717 DOI: 10.1155/2021/6139308] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Phenolic compounds are naturally present as secondary metabolites in plant-based sources such as fruits, vegetables, and spices. They have received considerable attention for their antioxidant, anti-inflammatory, and anti-carcinogenic properties for protection against many chronic disorders such as neurodegenerative diseases, diabetes, cardiovascular diseases, and cancer. They are categorized into various groups based on their chemical structure and include phenolic acids, flavonoids, curcumins, tannins, and quinolones. Their structural variations contribute to their specific beneficial effects on human health. The antioxidant property of phenolic compounds protects against oxidative stress by up-regulation of endogenous antioxidants, scavenging free radicals, and anti-apoptotic activity. Protocatechuic acid (PCA; 3,4-dihydroxy benzoic acid) and protocatechuic aldehyde (PAL; 3,4-dihydroxybenzaldehyde) are naturally occurring polyphenols found in vegetables, fruits, and herbs. PCA and PAL are the primary metabolites of anthocyanins and proanthocyanidins, which have been shown to possess pharmacological actions including antioxidant activity in vitro and in vivo. This review aims to explore the therapeutic potential of PCA and PAL by comprehensively summarizing their pharmacological properties reported to date, with an emphasis on their mechanisms of action and biological properties.
Collapse
Affiliation(s)
- Shijun Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhibo Gai
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ting Gui
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Juanli Chen
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Qingfa Chen
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- The Third Department of Cardiovascular Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
10
|
Zeng M, Shao C, Zhou H, He Y, Li W, Zeng J, Zhao X, Yang J, Wan H. Protocatechudehyde improves mitochondrial energy metabolism through the HIF1α/PDK1 signaling pathway to mitigate ischemic stroke-elicited internal capsule injury. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114232. [PMID: 34044078 DOI: 10.1016/j.jep.2021.114232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The internal capsule is vulnerable to ischemia, and mild ischemic stroke often results in lesion of the internal capsule, manifested as contralateral hemiplegia. Protocatechudehyde (PCA), a potential neuroprotective agent, has shown therapeutic effects in the study of a variety of nervous system diseases, including ischemic stroke. AIM OF THE STUDY The aim of this study was to evaluate the effects of PCA on cerebral ischemia reperfusion (CI/R)-elicited internal capsule injury and to elucidate the role of mitochondrial energy metabolism in the underlying mechanism of neuroprotective effects on ischemic stroke. MATERIALS AND METHODS A rat tMCAO model was established to investigate the therapeutic effects of intravenous PCA (20, 40, and 80 mg/kg, once per day, continued for 7 days) on CI/R-induced internal capsule injury and the regulation of PCA on molecules related to mitochondrial energy metabolism. In vitro, an OGD/R model of PC12 cells was established to further verify the therapeutic mechanism of PCA. RESULTS Results showed that PCA dose-dependently attenuated neurological deficit, reduced cerebral infarction, alleviated histopathological damage, and improved mitochondrial ultrastructure of the internal capsule after CI/R. Moreover, PCA reversed the upregulation of HIF1α, PDK1 and pPDHA1 expression induced by CI/R and significantly increased the content of acetyl-CoA, ATP, and the activity of ATP synthase. In vitro, PCA treatment promoted cell survival, inhibited apoptosis, attenuated the dissipation of mitochondrial membrane potential in OGD/R-treated PC12 cells, and these therapeutic effects were reversed by the combination of cobalt chloride (CoCl2), a specific pharmacological inducer of HIF1a expression. CONCLUSIONS These results indicate that PCA exerts a protective effect against CI/R-induced internal capsule injury and improves mitochondrial energy metabolism in the internal capsule, and the mechanism is associated with the inhibition of HIF1α/PDK1 signaling pathway.
Collapse
Affiliation(s)
- Miaolin Zeng
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Chongyu Shao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Huifen Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Wentao Li
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Jieqiong Zeng
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Xixi Zhao
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Jiehong Yang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China.
| | - Haitong Wan
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China; College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China.
| |
Collapse
|
11
|
Chang YT, Chung MC, Hsieh CC, Shieh JJ, Wu MJ. Evaluation of the Therapeutic Effects of Protocatechuic Aldehyde in Diabetic Nephropathy. Toxins (Basel) 2021; 13:toxins13080560. [PMID: 34437430 PMCID: PMC8402415 DOI: 10.3390/toxins13080560] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/30/2021] [Accepted: 08/08/2021] [Indexed: 11/18/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the most severe chronic kidney diseases in diabetes and is the main cause of end-stage renal disease (ESRD). Protocatechuic aldehyde (PCA) is a natural product with a variety of effects on pulmonary fibrosis. In this study, we examined the effects of PCA in C57BL/KS db/db male mice. Kidney morphology, renal function indicators, and Western blot, immunohistochemistry, and hematoxylin and eosin (H&E) staining data were analyzed. The results revealed that treatment with PCA could reduce diabetic-induced renal dysfunction, as indicated by the urine albumin-to-creatinine ratio (db/m: 120.1 ± 46.1μg/mg, db/db: 453.8 ± 78.7 µg/mg, db/db + 30 mg/kg PCA: 196.6 ± 52.9 µg/mg, db/db + 60 mg/kg PCA: 163.3 ± 24.6 μg/mg, p < 0.001). However, PCA did not decrease body weight, fasting plasma glucose, or food and water intake in db/db mice. H&E staining data revealed that PCA reduced glomerular size in db/db mice (db/m: 3506.3 ± 789.3 μm2, db/db: 6538.5 ± 1818.6 μm2, db/db + 30 mg/kg PCA: 4916.9 ± 1149.6 μm2, db/db + 60 mg/kg PCA: 4160.4 ± 1186.5 μm2p < 0.001). Western blot and immunohistochemistry staining indicated that PCA restored the normal levels of diabetes-induced fibrosis markers, such as transforming growth factor-beta (TGF-β) and type IV collagen. Similar results were observed for epithelial–mesenchymal transition-related markers, including fibronectin, E-cadherin, and α-smooth muscle actin (α-SMA). PCA also decreased oxidative stress and inflammation in the kidney of db/db mice. This research provides a foundation for using PCA as an alternative therapy for DN in the future.
Collapse
Affiliation(s)
- Yu-Teng Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402, Taiwan;
| | - Mu-Chi Chung
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 413, Taiwan
| | - Chang-Chi Hsieh
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan;
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402, Taiwan;
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Correspondence: (J.-J.S.); (M.-J.W.); Tel.: +886-4-23592525 (ext. 4052) (J.-J.S.); +886-4-23592525 (ext. 3000) (M.-J.W.)
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Graduate Institute of Clinical Medical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: (J.-J.S.); (M.-J.W.); Tel.: +886-4-23592525 (ext. 4052) (J.-J.S.); +886-4-23592525 (ext. 3000) (M.-J.W.)
| |
Collapse
|
12
|
Chen C, Chen J, Tao X, Fu M, Cheng B, Chen X. Activation of GPR30 with G1 inhibits oscillatory shear stress-induced adhesion of THP-1 monocytes to HAECs by increasing KLF2. Aging (Albany NY) 2021; 13:11942-11953. [PMID: 33875621 PMCID: PMC8109116 DOI: 10.18632/aging.202897] [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: 06/26/2020] [Accepted: 10/31/2020] [Indexed: 12/30/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease known to be mediated by numerous factors, among which endothelial dysfunction plays a critical role. Oscillatory shear stress induces endothelial cells to lose their anti-atherosclerotic properties and downregulates the expression of the innate protective transcription factor, Krüppel-like factor 2 (KLF2), which is typically upregulated in vascular endothelial cells in response to harmful stimuli. Oxidative stress and inflammation impair endothelial function and damage their survival. Oscillatory shear stress also promotes generation of reactive oxygen species and production of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), thereby further promoting endothelial dysfunction and formation of atherosclerotic plaque. A major event in the development of atherosclerotic plaque is rolling and adhesion of monocytes to endothelial cells, which is mediated by adhesion molecules including vascular cellular adhesion molecule 1 and endothelial-selectin. Expression of these molecules is also upregulated by oscillatory shear stress. Estrogen has long been recognized as a protective agent against atherosclerosis, but the mechanisms through which estrogen receptors prevent atherogenesis remain unclear. In the present study, we investigated the role of the G-coupled protein estrogen receptor (GPR30) in oscillatory shear stress- induced endothelial dysfunction. We show that agonism of GPR30 by its specific agonist G1 prevented oscillatory shear stress -induced oxidative stress markers and production of inflammatory cytokines and adhesion molecules. As a result, GPR30 activation suppresses monocytes adhesion to endothelial cells. Furthermore, we demonstrate that GPR30 prevents oscillatory shear stress- induced downregulation of KLF2 via ERK5 pathway. These findings suggest that endothelial GPR30 is potential target to suppress oscillatory shear stress mediated atherogenesis.
Collapse
Affiliation(s)
- Chi Chen
- Department of Geriatrics, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu 610072, Sichuang, China
| | - Jingyan Chen
- Department of Geriatrics, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu 610072, Sichuang, China
| | - Xuefei Tao
- Department of Geriatrics, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu 610072, Sichuang, China
| | - Minghuan Fu
- Department of Geriatrics, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu 610072, Sichuang, China
| | - Biao Cheng
- Department of Geriatrics, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu 610072, Sichuang, China
| | - Xiaohan Chen
- Department of Geriatrics, Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu 610072, Sichuang, China
| |
Collapse
|
13
|
Wan YJ, Wang YH, Guo Q, Jiang Y, Tu PF, Zeng KW. Protocatechualdehyde protects oxygen-glucose deprivation/reoxygenation-induced myocardial injury via inhibiting PERK/ATF6α/IRE1α pathway. Eur J Pharmacol 2021; 891:173723. [PMID: 33159933 DOI: 10.1016/j.ejphar.2020.173723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 01/31/2023]
Abstract
Endoplasmic reticulum (ER) stress has been considered as a promising strategy in developing novel therapeutic agents for cardiovascular diseases through inhibiting cardiomyocyte apoptosis. Protocatechualdehyde (PCA) is a natural phenolic compound from medicinal plant Salvia miltiorrhiza with cardiomyocyte protection. However, the potential mechanism of PCA on cardiovascular ischemic injury is largely unexplored. Here, we found that PCA exerted markedly anti-apoptotic effect in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced H9c2 cells (Rat embryonic ventricular H9c2 cardiomyocytes), which was detected by 3-(4, 5-dimethyl thiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT), lactate dehydrogenase (LDH), Hoechst 33258 and acridine orange/ethidium bromide (AO/EB) assays. PCA also obviously protected cardiomyocytes in myocardial fibrosis model of mice, which was determined by hematoxylin-eosin (HE) staining and TdT-mediated dUTP Nick-End Labeling (TUNEL) staining. Transcriptomics coupled with bioinformatics analysis revealed a complex pharmacological signaling network especially for PCA-mediated ER stress on cardiomyocytes. Further mechanism study suggested that PCA suppressed ER stress via inhibiting protein kinase R-like ER kinase (PERK), inositol-requiring enzyme1α (IRE1α), and transcription factor 6α (ATF6α) signaling pathway through Western blot, DIOC6 and ER-Tracker Red staining, leading to a protective effect against ER stress-mediated cardiomyocyte apoptosis. Taken together, our observations suggest that PCA is a major component from Salvia miltiorrhiza against cardiovascular ischemic injury by suppressing ER stress-associated PERK, IRE1α and ATF6α signaling pathways.
Collapse
Affiliation(s)
- Yan-Jun Wan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yan-Hang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
14
|
Fredette NC, Malik E, Mukhtar ML, Prossnitz ER, Terada N. A hypertension patient-derived iPSC model demonstrates a role for G protein-coupled estrogen receptor in hypertension risk and development. Am J Physiol Cell Physiol 2020; 319:C825-C838. [PMID: 32783656 DOI: 10.1152/ajpcell.00350.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypertension (HTN) is a polyfactorial disease that can manifest severe cardiovascular pathologies such as heart failure or stroke. Genome-wide association studies (GWAS) of HTN indicate that single-nucleotide polymorphisms (SNPs) contribute to increased risk for HTN and resistance to some HTN drug regimens (Hiltunen TP et al., J Am Heart Assoc 4: e001521, 2015; Le MT et al., PLoS One 8: e52062, 2013; McDonough CW et al., J Hypertens 31: 698-704, 2013; Vandell AG et al., Hypertension 60: 957-964, 2012). However, cellular mechanistic insights of such SNPs remain largely unknown. Using a bank of induced pluripotent stem cells (iPSCs) derived from patients with HTN and CRISPR/Cas9-mediated gene-editing approach, we investigated the effects of a female HTN risk-associated SNP (rs1154431) of the G protein-coupled estrogen receptor (GPER) (Bassuk SS, Manson JE., Clin Chem 60: 68-77, 2014) in vascular endothelial cells. Although GPER1 deletion reduced endothelial nitric oxide synthase (eNOS) activation in iPSC-derived endothelial cells (iECs), the polymorphism itself did not significantly affect eNOS and NO production in a comparison of isogenic hemizygous iECs expressing either normal (P16) or HTN-associated (L16) GPER. Interestingly, we demonstrate for the first time that GPER plays a role in regulation of adhesion molecule expression and monocyte adhesion to iECs. Moreover, the L16 iECs had higher expression of inflammation genes than P16 iECs, implying that the risk variant may affect carrier individuals through increased inflammatory activity. This study further indicates that iPSCs are a useful platform for exploring mechanistic insights underlying hypertension GWAS endeavors.
Collapse
Affiliation(s)
- Natalie C Fredette
- Department of Pathology, Immunology and Experimental Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Eliyah Malik
- Department of Pathology, Immunology and Experimental Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Marah L Mukhtar
- Department of Mechanical & Aerospace Engineering, University of Florida Herbert Wertheim College of Engineering, Gainesville, Florida
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Naohiro Terada
- Department of Pathology, Immunology and Experimental Medicine, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
15
|
Li F, Zhang T, He Y, Gu W, Yang X, Zhao R, Yu J. Inflammation inhibition and gut microbiota regulation by TSG to combat atherosclerosis in ApoE -/- mice. JOURNAL OF ETHNOPHARMACOLOGY 2020; 247:112232. [PMID: 31606534 DOI: 10.1016/j.jep.2019.112232] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/20/2019] [Accepted: 09/11/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 2,3,5,4'-Tetrahydroxy-stilbene-2-O-β-D-glucoside (TSG) is the main active component of Polygoni Multiflori Radix, a root of the homonymous plant widely used in traditional Chinese medicine. TSG has protective effects on the liver, reduces cholesterol and possesses anti-oxidant, anti-tumor, and anti-atherosclerotic properties. However, the pharmacological effects and mechanisms of action of Polygonum multiflorum on atherosclerosis (AS) have not been studied yet. PURPOSE The aim of this research was to study the effects of Polygoni Multiflori Radix Praeparata (PMRP) and its major active chemical constituent TSG on AS in ApoE-deficient (ApoE-/-) mice fed with high fat diets to provide a scientific basis in the use of PMRP and TSG against cardiovascular diseases. METHODS High fat diet induced AS in ApoE-/- mice were treated with PMRP, TSG (low and high doses), and simvastatin (SIM) for 8 weeks. At the end of the treatment, mouse serum lipid levels, triglycerides (TG), and total cholesterol (TC) were measured by an oxidase method (other indicators were determined by ELISA), while the content in oxidized low density lipoprotein (ox-LDL) and the expression of inflammatory factors such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), vascular cell adhesion molecule-1 (VCAM-1), and monocyte chemotactic protein-1 (MCP-1) in the serum and aortic samples were measured by ELISA. Atherosclerotic plaque morphology was evaluated by oil red O in thoracic aorta. In addition, 16S rDNA-V4 hypervariable region genome sequence of all microbes in the fecal sample from each group was analyzed to evaluate potential structure changes in the gut microbiota after treatment with PMRP and TSG. RESULTS TSG markedly inhibited AS plaque formation in ApoE-/- mice. Furthermore, PMRP and TSG improved lipid accumulation by reducing TG and ox-LDL levels. TSG inhibited inflammation by the down-regulation of IL-6, TNF-α, VCAM-1 and MCP-1 expression in serum, and PMRP inhibited inflammation by reducing VCAM-1, ICAM-1 and CCRA expression in aortic tissue. In addition, TSG reduced or prevented AS by the regulation of the composition of the overall gut microbiota, such as Firmicutes, Bacteroidetes, Tenericutes, Proteobacteria phyla, Akkermensia genera and Helicobacter pylori. CONCLUSION PMRP and TSG improved lipid accumulation and inflammation, and regulated the intestinal microbial imbalance in ApoE-/- mice. TSG exerted a preventive effect in the development and progression of AS.
Collapse
Affiliation(s)
- Fengjiao Li
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Ting Zhang
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Yanran He
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Wen Gu
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Xingxin Yang
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Ronghua Zhao
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Jie Yu
- Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China.
| |
Collapse
|
16
|
Notas G, Kampa M, Castanas E. G Protein-Coupled Estrogen Receptor in Immune Cells and Its Role in Immune-Related Diseases. Front Endocrinol (Lausanne) 2020; 11:579420. [PMID: 33133022 PMCID: PMC7564022 DOI: 10.3389/fendo.2020.579420] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/10/2020] [Indexed: 12/30/2022] Open
Abstract
G protein-coupled estrogen receptor 1 (GPER1), is a functional estrogen receptor involved in estrogen related actions on several systems including processes of the nervous, reproductive, metabolic, cardiovascular, and immune system. Regarding the latter, GPER is expressed in peripheral B and T lymphocytes as well as in monocytes, eosinophils, and neutrophils. Several studies have implicated GPER in immune-mediated diseases like multiple sclerosis, Parkinson's disease, and atherosclerosis-related inflammation, while a recent report suggests that its deletion could be responsible for a form of familial immunodeficiency. It has also been suggested that it is a key regulator of immune-mediated events in breast, pancreatic, prostate, and hepatocellular cancer as well as in melanoma. GPER has been also reported to interact with classic ER-alpha or its splice variants in order to modify immune functions. This review aims to present current knowledge relating GPER to immune functions, the cellular and signaling pathways involved, as well as the potential clinical implications of GPER modulation in immune-related diseases.
Collapse
|
17
|
Imam Aliagan A, Madungwe NB, Tombo N, Feng Y, Bopassa JC. Chronic GPER1 Activation Protects Against Oxidative Stress-Induced Cardiomyoblast Death via Preservation of Mitochondrial Integrity and Deactivation of Mammalian Sterile-20-Like Kinase/Yes-Associated Protein Pathway. Front Endocrinol (Lausanne) 2020; 11:579161. [PMID: 33193095 PMCID: PMC7604496 DOI: 10.3389/fendo.2020.579161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction: Estrogen (17β-estradiol, E2) is well-known to induce cardioprotective effects against ischemia/reperfusion (I/R) injury. We recently reported that acute application of E2 at the onset of reperfusion in vivo induces cardioprotective effects against I/R injury via activation of its non-steroidal receptor, G protein-coupled estrogen receptor 1 (GPER1). Here, we investigated the impact and mechanism underlying chronic GPER1 activation in cultured H9c2 rat cardiomyoblasts. Methods: H9c2 rat cardiomyoblasts were cultured and pretreated with the cytotoxic agent H2O2 for 24 h and incubated in the presence of vehicle (control), GPER1 agonists E2 and G1, or GPER1 agonists supplemented with G15 (GPER1 antagonist) for 48 or 96 h. After treatment, cells were collected to measure the rate of cell death and viability using flow cytometry and Calcein AM assay or MTT assay, respectively. The resistance to opening of the mitochondrial permeability transition pore (mPTP), the mitochondrial membrane potential, and ATP production was assessed using fluorescence microscopy, and the mitochondrial structural integrity was observed with electron microscopy. The levels of the phosphorylation of mammalian sterile-20-like kinase (MST1) and yes-associated protein (YAP) were assessed by Western blot analysis in whole-cell lysate, while the expression levels of mitochondrial biogenesis genes, YAP target genes, and proapoptotic genes were measured by qRT-PCR. Results: We found that after H2O2 treatment, chronic E2/G1 treatment decreased cell death effect was associated with the prevention of the S phase of the cell cycle arrest compared to control. In the mitochondria, chronic E2/G1 activation treatment preserved the cristae morphology, and increased resistance to opening of mPTP, but with little change to mitochondrial fusion/fission. Additionally, chronic E2/G1 treatment predominantly reduced phosphorylation of MST1 and YAP, as well as increased MST1 and YAP protein levels. E2 treatment also upregulated the expression levels of TGF-β and PGC-1α mRNAs and downregulated PUMA and Bim mRNAs. Except for ATP production, all the E2 or G1 effects were prevented by the cotreatment with the GPER1 antagonist, G15. Conclusion: Together, these results indicate that chronic GPER1 activation with its agonists E2 or G1 treatment protects H9c2 cardiomyoblasts against oxidative stress-induced cell death and increases cell viability by preserving mitochondrial structure and function as well as delaying the opening of mPTP. These chronic GPER1 effects are associated with the deactivation of the non-canonical MST1/YAP mechanism that leads to genetic upregulation of cell growth genes (CTGF, CYR61, PGC-1α, and ANKRD1), and downregulation of proapoptotic genes (PUMA and Bim).
Collapse
Affiliation(s)
- Abdulhafiz Imam Aliagan
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ngonidzashe B. Madungwe
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Nathalie Tombo
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Yansheng Feng
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Jean C. Bopassa
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- *Correspondence: Jean C. Bopassa
| |
Collapse
|
18
|
Yang G, Zhuo J, Lin Y, Zhang M, Liu L, Chen X, Gao R. Ginsenoside Rb1 Prevents Dysfunction of Endothelial Cells by Suppressing Inflammatory Response and Apoptosis in the High-Fat Diet Plus Balloon Catheter-Injured Rabbit Model via the G Protein-Coupled Estrogen Receptor-Mediated Phosphatidylinositol 3-Kinases (PI3K)/Akt Pathway. Med Sci Monit 2019; 25:7407-7417. [PMID: 31609302 PMCID: PMC6791186 DOI: 10.12659/msm.912986] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/28/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The initiation of atherosclerosis (AS) is attributed to the dysfunction of endothelial cells (ECs) via the inhibition of g protein-coupled estrogen receptor (GPER). In the current study, we assessed the potential of Ginsenoside Rb1 (Rb1) to attenuate the dysfunction of ECs via GPER-mediated PI3K/Akt pathway. MATERIAL AND METHODS AS was induced in rabbits and then the AS rabbits were treated with Rb1. Thereafter, the ECs were isolated from AS and healthy rabbits, and treated with Rb1. The effect of Rb1 on blood lipid levels in AS rabbits and on apoptosis, inflammatory response, and GPER/PI3K/Akt axis activity in ECs was detected. Furthermore, the activities of GPER and PI3K were modulated to verify the key role of the axis in the anti-AS effect of Rb1. RESULTS The levels of total cholesterol, low-density lipoprotein (LDL), and triglyceride in AS rabbits were suppressed by Rb1 while the high-density lipoprotein (HDL) level was increased. In in vitro assays, Rb1 administration inhibited apoptosis process and the production of pro-inflammation cytokines in AS ECs. The expression levels of GPER, p-PI3K, and p-Akt were upregulated by Rb1, associated with the increased level of Bcl-2 and reduced level of Bax. When the activity of GPER was inhibited by GP-15 in AS ECs, the treatment effect of Rb1 was blocked. However, the activation of PI3K could restore the protective effect of Rb1 after the inhibition of GPER. CONCLUSIONS The anti-AS potential of Rb1 was exerted by restoring the regular function of ECs via the activation of GPER-mediated PI3K/Akt signaling.
Collapse
Affiliation(s)
- Guoliang Yang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| | - Jun Zhuo
- Department of Interventional Radiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| | - Yuedong Lin
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| | - Meng Zhang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| | - Lixin Liu
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| | - Xueying Chen
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| | - Ronghua Gao
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, P.R. China
| |
Collapse
|
19
|
Davison AS, Norman BP, Ross GA, Hughes AT, Khedr M, Milan AM, Gallagher JA, Ranganath LR. Evaluation of the serum metabolome of patients with alkaptonuria before and after two years of treatment with nitisinone using LC-QTOF-MS. JIMD Rep 2019; 48:67-74. [PMID: 31392115 PMCID: PMC6606987 DOI: 10.1002/jmd2.12042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/26/2019] [Accepted: 05/02/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The homogentisic acid-lowering therapy nitisinone is being evaluated for the treatment of alkaptonuria (AKU) at the National Centre for AKU. Beyond hypertyrosinemia, the wider metabolic consequences of its use are largely unknown. The aim of this work was to evaluate the impact of nitisinone on the serum metabolome of patients with AKU after 12 and 24 months of treatment. METHODS Deproteinized serum from 25 patients with AKU (mean age[±SD] 51.1 ± 14.9 years, 12 male) was analyzed using the 1290 Infinity II liquid chromatography system coupled to a 6550 quadrupole time-of-flight mass spectrometry (Agilent, UK). Raw data were processed using a batch targeted feature extraction algorithm and an accurate mass retention time database containing 469 intermediary metabolites (MW 72-785). Matched entities (±10 ppm theoretical accurate mass and ±0.3 minutes retention time window) were filtered based on their frequency and variability (<25% CV) in group quality control samples, and repeated measures statistical significance analysis with Benjamini-Hochberg false discovery rate adjustment was used to assess changes in metabolite abundance. RESULTS Eight metabolites increased in abundance (log2 fold change [FC] 2.1-15.2, P < .05); 7 of 8 entities were related to tyrosine metabolism, and 13 decreased in abundance (log2 FC 1.5-15.5, P < .05); including entities related to tyrosine (n = 2), tryptophan (n = 3), xanthine (n = 2), and citric acid cycle metabolism (n = 2). CONCLUSIONS Evaluation of the serum metabolome of patients with AKU showed a significant difference in the abundance of several metabolites following treatment with nitisinone, including a number that have not been previously reported; several of these were not related to the tyrosine metabolic pathway. SYNOPSIS Nitisinone therapy has a significant impact on several metabolites beyond the tyrosine metabolic pathway, several of which appear to be related to the redox state of the cell.
Collapse
Affiliation(s)
- Andrew S. Davison
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University Hospitals TrustLiverpoolUK
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| | - Brendan P. Norman
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| | | | - Andrew T. Hughes
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University Hospitals TrustLiverpoolUK
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| | - Milad Khedr
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University Hospitals TrustLiverpoolUK
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| | - Anna M. Milan
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University Hospitals TrustLiverpoolUK
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| | - James A. Gallagher
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| | - Lakshminarayan R. Ranganath
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University Hospitals TrustLiverpoolUK
- Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of Liverpool, Liverpool Health PartnersLiverpoolUK
| |
Collapse
|
20
|
Yang F, Lin ZW, Huang TY, Chen TT, Cui J, Li MY, Hua YQ. Ligustilide, a major bioactive component of Angelica sinensis, promotes bone formation via the GPR30/EGFR pathway. Sci Rep 2019; 9:6991. [PMID: 31061445 PMCID: PMC6502875 DOI: 10.1038/s41598-019-43518-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022] Open
Abstract
Angelica sinensis (Oliv.) Diels is a widely-used traditional Chinese herbal medicine in treating osteoporosis. Ligustilide (LIG) is the main component of A. sinensis and is considered to be the most effective biologically active ingredient in this plant. LIG has been found to have multiple pharmacological activities, such as anti-atherosclerosis, neuroprotection, anticancer, anti-inflammatory and analgesic. However, little is known regarding its anti-osteoporotic effects. The aims of this study were to investigate any protective effect of LIG on bone formation. The results showed that LIG significantly ameliorated inhibition of bone formation in zebrafish caused by prednisolone. LIG promoted osteoblast differentiation, including that of the pre-osteoblastic cell line MC3T3-E1 and bone marrow mesenchymal stem cells. LIG greatly improved the viability of MC3T3-E1 cells exposed to H2O2, attenuated H2O2-induced apoptosis and increased the expression of Bcl-2. Furthermore, LIG treatment lead to marked activation of phosphorylated EGFR and ERK1/2. These effects could be obviously inhibited by blocking GPR30 signaling with the specific inhibitor G15. Collectively, the results reveal that GPR30 is a positive switch for LIG to increase bone formation via regulation of EGFR, and these results provide evidence for the potential of LIG to treat osteoporosis.
Collapse
Affiliation(s)
- F Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Z W Lin
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - T Y Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - T T Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - J Cui
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - M Y Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Y Q Hua
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China.
| |
Collapse
|
21
|
Peixoto P, da Silva JF, Aires RD, Costa ED, Lemos VS, Bissoli NS, dos Santos RL. Sex difference in GPER expression does not change vascular relaxation or reactive oxygen species generation in rat mesenteric resistance arteries. Life Sci 2018; 211:198-205. [DOI: 10.1016/j.lfs.2018.09.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/29/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023]
|
22
|
Wnuk A, Rzemieniec J, Litwa E, Lasoń W, Kajta M. Prenatal exposure to benzophenone-3 (BP-3) induces apoptosis, disrupts estrogen receptor expression and alters the epigenetic status of mouse neurons. J Steroid Biochem Mol Biol 2018; 182:106-118. [PMID: 29704544 DOI: 10.1016/j.jsbmb.2018.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
Current evidence indicates that benzophenone-3 (BP-3) can pass through the placental and blood-brain barriers and thus can likely affect infant neurodevelopment. Despite widespread exposure, data showing the effects of BP-3 on the developing nervous system are scarce. This study revealed for the first time that prenatal exposure to BP-3 led to apoptosis and neurotoxicity, altered the levels of estrogen receptors (ERs) and changed the epigenetic status of mouse neurons. In the present study, subcutaneous injections of pregnant mice with BP-3 at 50 mg/kg, which is an environmentally relevant dose, evoked activation of caspase-3 and lactate dehydrogenase (LDH) release as well as substantial loss of mitochondrial membrane potential in neocortical cells of their embryonic offspring. Apoptosis-focused microarray analysis of neocortical cells revealed up-regulation of 22 genes involved in apoptotic cell death. This effect was supported by increased BAX and CASP3 mRNA and protein levels, as evidenced by qPCR, ELISAs and western blots. BP-3-induced apoptosis and neurotoxicity were accompanied by decreases in the mRNA and protein expression levels of ESR1 and ESR2 (also known as ERα and ERβ), with a simultaneous increase in GPER1 (also known as GPR30) expression. In addition to the demonstration that treatment of pregnant mice with BP-3 induced apoptosis, caused neurotoxicity and altered ERs expression levels in neocortical cells of their embryonic offspring, we showed that prenatal administration of BP-3 inhibited global DNA methylation as well as reduced DNMTs activity. BP-3 also caused specific hypomethylation of the genes Gper1 and Bax, an effect that was accompanied by increased mRNA and protein expression levels. In addition, BP-3 caused hypermethylation of the genes Esr1, Esr2 and Bcl2, which could explain the reduced mRNA and protein levels of the estrogen receptors. This study demonstrated for the first time that prenatal exposure to BP-3 caused severe neuronal apoptosis that was accompanied by impaired ESR1/ESR2 expression, enhanced GPER1 expression, global DNA hypomethylation and altered methylation statuses of apoptosis-related and ERs genes. We suggest that the effects of BP-3 in embryonic neurons may be the fetal basis of the adult onset of nervous system disease.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Joanna Rzemieniec
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Ewa Litwa
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Władysław Lasoń
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Małgorzata Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland.
| |
Collapse
|
23
|
Li ZM, Xu SW, Liu PQ. Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharmacol Sin 2018; 39:802-824. [PMID: 29698387 PMCID: PMC5943903 DOI: 10.1038/aps.2017.193] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 12/31/2017] [Indexed: 02/07/2023] Open
Abstract
Salvia miltiorrhiza Burge (Danshen) is an eminent medicinal herb that possesses broad cardiovascular and cerebrovascular protective actions and has been used in Asian countries for many centuries. Accumulating evidence suggests that Danshen and its components prevent vascular diseases, in particular, atherosclerosis and cardiac diseases, including myocardial infarction, myocardial ischemia/reperfusion injury, arrhythmia, cardiac hypertrophy and cardiac fibrosis. The published literature indicates that lipophilic constituents (tanshinone I, tanshinone IIa, tanshinone IIb, cryptotanshinone, dihydrotanshinone, etc) as well as hydrophilic constituents (danshensu, salvianolic acid A and B, protocatechuic aldehyde, etc) contribute to the cardiovascular protective actions of Danshen, suggesting a potential synergism among these constituents. Herein, we provide a systematic up-to-date review on the cardiovascular actions and therapeutic potential of major pharmacologically active constituents of Danshen. These bioactive compounds will serve as excellent drug candidates in small-molecule cardiovascular drug discovery. This article also provides a scientific rationale for understanding the traditional use of Danshen in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Zhuo-ming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510006, China
| | - Suo-wen Xu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Pei-qing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
24
|
4-Hydroxybenzaldehyde accelerates acute wound healing through activation of focal adhesion signalling in keratinocytes. Sci Rep 2017; 7:14192. [PMID: 29079748 PMCID: PMC5660242 DOI: 10.1038/s41598-017-14368-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/09/2017] [Indexed: 01/16/2023] Open
Abstract
4-Hydroxybenzaldehyde (4-HBA) is a naturally occurring benzaldehyde and the major active constituent of Gastrodia elata. While recent studies have demonstrated metabolic effects of 4-HBA, little is known about the physiological role of 4-HBA in acute wound healing. Here, we investigated the effects and mechanisms of 4-HBA on acute wound healing. Using an in vitro approach, we found that 4-HBA significantly promoted keratinocyte cell migration and invasion by increasing focal adhesion kinase and Src activity. In addition, 4-HBA treatment also promoted wound healing and re-epithelialization in an in vivo excision wound animal model. Combination treatment with 4-HBA and platelet-derived growth factor subunit B homodimer showed synergistic effects in promoting wound healing. Taken together, our results demonstrated that treatment with 4-HBA promoted keratinocyte migration and wound healing in mouse skin through the Src/mitogen-activated protein kinase pathway. Therefore, 4-HBA could be a candidate therapeutic agent with the potential to promote acute wound healing.
Collapse
|
25
|
Wang X, He X, Deng X, He Y, Zhou X. Roles of miR‑4463 in H2O2‑induced oxidative stress in human umbilical vein endothelial cells. Mol Med Rep 2017; 16:3242-3252. [PMID: 28713907 PMCID: PMC5547966 DOI: 10.3892/mmr.2017.7001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 04/28/2017] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is implicated in the pathophysiology of vascular diseases, including atherosclerosis, aneurysm and arteriovenous fistula. A previous study from our lab suggested that microRNA (miR)-4463 may be involved in the pathogenesis of vascular disease; however, the roles of oxidative stress in the molecular mechanisms underlying the actions of miR-4463 in vascular disease have yet to be elucidated. The aim of the present study was to investigate the role of miR-4463 in hydrogen peroxide (H2O2)-induced oxidative stress in human umbilical vein endothelial cells (HUVECs). Reverse transcription-quantitative polymerase chain reaction was used to assess the expression levels of miR-4463 in HUVECs treated with various concentrations of H2O2. Flow cytometry was used to evaluate the percentage of apoptotic cells, and the protein expression levels of the apoptotic markers cleaved (C)-caspase3, poly (adenosine diphosphate-ribose) polymerase 1 (PARP1), B cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax) and X-linked inhibitor of apoptosis protein (XIAP) were determined using western blot analysis. The results demonstrated that the apoptotic rate of HUVECs was increased following treatment with H2O2 in a concentration-dependent manner, and the expression of miR-4463 was also upregulated in a dose-dependent manner. Following transfection with miR-4463 mimics, the levels of malondialdehyde and reactive oxygen species were increased in HUVECs, with a corresponding increase in the apoptotic rate. Furthermore, western blot analysis revealed that the protein expression levels of C-caspase3, PARP1 and Bax were upregulated, whereas the levels of Bcl-2 and XIAP were downregulated. In conclusion, the present findings suggested that the upregulation of miR-4463 may enhance H2O2-induced oxidative stress and promote apoptosis in HUVECs in vitro.
Collapse
Affiliation(s)
- Xueqin Wang
- Department of Vascular and Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Jiangyang, Luzhou, Sichuan 646000, P.R. China
| | - Xuemei He
- Medical Research Center, The Affiliated Hospital of Southwest Medical University, Jiangyang, Luzhou, Sichuan 646000, P.R. China
| | - Xian Deng
- Department of Vascular and Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Jiangyang, Luzhou, Sichuan 646000, P.R. China
| | - Yanzheng He
- Department of Vascular and Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Jiangyang, Luzhou, Sichuan 646000, P.R. China
| | - Xiangyu Zhou
- Department of Vascular and Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Jiangyang, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
26
|
Kong BS, Im SJ, Lee YJ, Cho YH, Do YR, Byun JW, Ku CR, Lee EJ. Vasculoprotective Effects of 3-Hydroxybenzaldehyde against VSMCs Proliferation and ECs Inflammation. PLoS One 2016; 11:e0149394. [PMID: 27002821 PMCID: PMC4803227 DOI: 10.1371/journal.pone.0149394] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/01/2016] [Indexed: 01/15/2023] Open
Abstract
3-hydroxybenzaldehyde (3-HBA) is a precursor compound for phenolic compounds like Protocatechuic aldehyde (PCA). From recent reports, PCA has shown vasculoprotective potency, but the effects of 3-HBA remain unclear. The aim of this study is to investigate the vasculoprotective effects of 3-HBA in endothelial cells, vascular smooth muscle cells and various animal models. We tested effects of 3-HBA in both vitro and vivo. 3-HBA showed that it prevents PDGF-induced vascular smooth muscle cells (VSMCs) migration and proliferation from MTS, BrdU assays and inhibition of AKT phosphorylation. It arrested S and G0/G1 phase of VSMC cell cycle in PI staining and it also showed inhibited expression levels of Rb1 and CD1. In human umbilical vein endothelial cells (HUVECs), 3-HBA inhibited inflammatory markers and signaling molecules (VCAM-1, ICAM-1, p-NF-κB and p-p38). For ex vivo, 3-HBA has shown dramatic effects in suppressing the sprouting from aortic ring of Spargue Dawley (SD) rats. In vivo data supported the vasculoprotective effects of 3-HBA as it inhibited angiogenesis from Matrigel Plug assay in C57BL6 mouse, prevented ADP-induced thrombus generation, increased blood circulation after formation of thrombus, and attenuated neointima formation induced by common carotid artery balloon injury of SD rats. 3-HBA, a novel therapeutic agent, has shown vasculoprotective potency in both in vitro and in vivo.
Collapse
MESH Headings
- Animals
- Benzaldehydes/pharmacology
- Carotid Artery Injuries/metabolism
- Catechols/pharmacology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- G1 Phase/drug effects
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Inflammation/drug therapy
- Inflammation/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- Neointima/drug therapy
- Neointima/metabolism
- Phosphorylation/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Sprague-Dawley
- Resting Phase, Cell Cycle/drug effects
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Byung Soo Kong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Soo Jung Im
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Yang Jong Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Yoon Hee Cho
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Yu Ri Do
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Jung Woo Byun
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Cheol Ryong Ku
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
| | - Eun Jig Lee
- Endocrinology, Institute of Endocrine Research, College of Medicine, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
27
|
Moreno-Ulloa A, Mendez-Luna D, Beltran-Partida E, Castillo C, Guevara G, Ramirez-Sanchez I, Correa-Basurto J, Ceballos G, Villarreal F. The effects of (-)-epicatechin on endothelial cells involve the G protein-coupled estrogen receptor (GPER). Pharmacol Res 2015; 100:309-20. [PMID: 26303816 DOI: 10.1016/j.phrs.2015.08.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 02/05/2023]
Abstract
We have provided evidence that the stimulatory effects of (-)-epicatechin ((-)-EPI) on endothelial cell nitric oxide (NO) production may involve the participation of a cell-surface receptor. Thus far, such entity(ies) has not been fully elucidated. The G protein-coupled estrogen receptor (GPER) is a cell-surface receptor that has been linked to protective effects on the cardiovascular system and activation of intracellular signaling pathways (including NO production) similar to those reported with (-)-EPI. In bovine coronary artery endothelial cells (BCAEC) by the use of confocal imaging, we evidence the presence of GPER at the cell-surface and on F-actin filaments. Using in silico studies we document the favorable binding mode between (-)-EPI and GPER. Such binding is comparable to that of the GPER agonist, G1. By the use of selective blockers, we demonstrate that the activation of ERK 1/2 and CaMKII by (-)-EPI is dependent on the GPER/c-SRC/EGFR axis mimicking those effects noted with G1. We also evidence by the use of siRNA the role that GPER has on mediating ERK1/2 activation by (-)-EPI. GPER appears to be coupled to a non Gαi/o or Gαs, protein subtype. To extrapolate our findings to an ex vivo model, we employed phenylephrine pre-contracted aortic rings evidencing that (-)-EPI can mediate vasodilation through GPER activation. In conclusion, we provide evidence that suggests the GPER as a potential mediator of (-)-EPI effects and highlights the important role that GPER may have on cardiovascular system protection.
Collapse
Affiliation(s)
- Aldo Moreno-Ulloa
- University of California, San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA, USA; Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - David Mendez-Luna
- Laboratorio de modelado Molecular y Diseño de Fármacos, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | | | - Carmen Castillo
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Gustavo Guevara
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Israel Ramirez-Sanchez
- University of California, San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA, USA; Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - José Correa-Basurto
- Laboratorio de modelado Molecular y Diseño de Fármacos, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico; Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Guillermo Ceballos
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Francisco Villarreal
- University of California, San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA, USA.
| |
Collapse
|
28
|
Citreoviridin Enhances Atherogenesis in Hypercholesterolemic ApoE-Deficient Mice via Upregulating Inflammation and Endothelial Dysfunction. PLoS One 2015; 10:e0125956. [PMID: 25933220 PMCID: PMC4416801 DOI: 10.1371/journal.pone.0125956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/27/2015] [Indexed: 11/19/2022] Open
Abstract
Vascular endothelial dysfunction and inflammatory response are early events during initiation and progression of atherosclerosis. In vitro studies have described that CIT markedly upregulates expressions of ICAM-1 and VCAM-1 of endothelial cells, which result from NF-κB activation induced by CIT. In order to determine whether it plays a role in atherogenesis in vivo, we conducted the study to investigate the effects of CIT on atherosclerotic plaque development and inflammatory response in apolipoprotein E deficient (apoE-/-) mice. Five-week-old apoE-/- mice were fed high-fat diets and treated with CIT for 15 weeks, followed by assay of atherosclerotic lesions. Nitric oxide (NO), vascular endothelial growth factor (VEGF) and endothelin-1 (ET-1) were detected in serum. Levels of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), VEGF, and ET-1 in plaque areas of artery walls were examined. NF-κB p65 expression and NF-κB activation in aorta also were assessed. CIT treatment significantly augmented atherosclerotic plaques and increased expressions of ICAM-1, VCAM-1, VEGF and ET-1 in aorta. Mechanistic studies showed that activation of NF-κB was significantly elevated by CIT treatment, indicating the effect of CIT on atherosclerosis may be regulated by activation of NF-κB.
Collapse
|