1
|
Tuo P, Zhao R, Li N, Yan S, Yang G, Wang C, Sun J, Sun H, Wang M. Lycorine inhibits Ang II-induced heart remodeling and inflammation by suppressing the PI3K-AKT/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155464. [PMID: 38484625 DOI: 10.1016/j.phymed.2024.155464] [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: 12/13/2023] [Revised: 02/03/2024] [Accepted: 02/16/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Ang II induces hypertensive heart failure (HF) via hemodynamic and non-hemodynamic actions. Lycorine (LYC) is an alkaloid derived from Lycoris bulbs, and it possesses anti-cardiovascular disease-related activities. Herein, we explored the potential LYC-mediated regulation of Ang II-induced HF. METHODS Over 4 weeks, we established a hypertensive HF mouse model by infusing Ang II into C57BL/6 mice using a micro-osmotic pump. For the final two weeks, mice were administered LYC via intraperitoneal injection. The LYC signaling network was then deduced using RNA sequencing. RESULTS LYC administration strongly suppressed hypertrophy, myocardial fibrosis, and cardiac inflammation. As a result, it minimized heart dysfunction while causing no changes in blood pressure. The Nuclear Factor kappa B (NF-κB) network/phosphoinositol-3-kinase (PI3K)-protein kinase B (AKT) was found to be a major modulator of LYC-based cardioprotection using RNA sequencing study. We further confirmed that in cultured cardiomyocytes and mouse hearts, LYC reduced the inflammatory response and downregulated the Ang II-induced PI3K-AKT/NF-κB network. Moreover, PI3K-AKT or NF-κB axis depletion in cardiomyocytes completely abrogated the anti-inflammatory activities of LYC. CONCLUSION Herein, we demonstrated that LYC safeguarded hearts in Ang II -stimulated mice by suppressing the PI3K-AKT/NF-κB-induced inflammatory responses. Given the evidence mentioned above, LYC is a robust therapeutic agent for hypertensive HF.
Collapse
Affiliation(s)
- Pingping Tuo
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China
| | - Risheng Zhao
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China
| | - Ning Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin, Changchun, 130012, China
| | - Shuang Yan
- Department of Ultrasonography, Inteqrated Traditional Chinese and Western Medicine Hospital of Jilin city Jilin Province, Jilin, 132000, China
| | - Gege Yang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China
| | - Chunmei Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China
| | - Jinghui Sun
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China
| | - Haiming Sun
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China.
| | - Mengyang Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin, 132000, China.
| |
Collapse
|
2
|
Niu Z, Li X, Yang X, Sun Z. Protective effects of sinomenine against dextran sulfate sodium-induced ulcerative colitis in rats via alteration of HO-1/Nrf2 and inflammatory pathway. Inflammopharmacology 2024; 32:2007-2022. [PMID: 38573363 DOI: 10.1007/s10787-024-01455-6] [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: 02/07/2024] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Dextran Sulfate Sodium (DSS) induces ulcerative colitis (UC), a type of inflammatory bowel disease (IBD) that leads to inflammation, swelling, and ulcers in the large intestine. The aim of this experimental study is to examine how sinomenine, a plant-derived alkaloid, can prevent or reduce the damage caused by DSS in the colon and rectum of rats. MATERIAL AND METHODS Induction of ulcerative colitis (UC) in rats was achieved by orally administering a 2% Dextran Sulfate Sodium (DSS) solution, while the rats concurrently received oral administrations of sinomenine and sulfasalazine. The food, water intake was estimated. The body weight, disease activity index (DAI), colon length and spleen index estimated. Antioxidant, cytokines, inflammatory parameters and mRNA expression were estimated. The composition of gut microbiota was analyzed at both the phylum and genus levels in the fecal samples obtained from all groups of rats. RESULTS Sinomenine treatment enhanced the body weight, colon length and reduced the DAI, spleen index. Sinomenine treatment remarkably suppressed the level of NO, MPO, ICAM-1, and VCAM-1 along with alteration of antioxidant parameters such as SOD, CAT, GPx, GR and MDA. Sinomenine treatment also decreased the cytokines like TNF-α, IL-1, IL-1β, IL-6, IL-10, IL-17, IL-18 in the serum and colon tissue; inflammatory parameters viz., PAF, COX-2, PGE2, iNOS, NF-κB; matrix metalloproteinases level such as MMP-1 and MMP-2. Sinomenine significantly (P < 0.001) enhanced the level of HO-1 and Nrf2. Sinomenine altered the mRNA expression of RIP1, RIP3, DRP3, NLRP3, IL-1β, caspase-1 and IL-18. Sinomenine remarkably altered the relative abundance of gut microbiota like firmicutes, Bacteroidetes, F/B ratio, Verrucomicrobia, and Actinobacteria. CONCLUSION The results clearly indicate that sinomenine demonstrated a protective effect against DSS-induced inflammation, potentially through the modulation of inflammatory pathways and gut microbiota.
Collapse
Affiliation(s)
- Zhongbao Niu
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xinhong Li
- Department of Outpatient Surgery, Central Hospital Affiliated to Shandong First Medical University, No. 105 Jiefang Road, Jinan, 250013, Jinan, China
| | - Xiuhua Yang
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, No. 105 Jiefang Road, Jinan, 250013, Jinan, China
| | - Zhongwei Sun
- Department of Gastrointestinal Surgery, Jinan Central Hospital, No.105, Jiefang Road, Lixia District, Jinan, 250013, Shandong, China.
| |
Collapse
|
3
|
Liu Y, Chen H, Wu Y, Ai F, Li W, Peng H, Gui F, Yu B, Chen Z. Sinomenine attenuates bleomycin-induced pulmonary fibrosis, inflammation, and oxidative stress by inhibiting TLR4/NLRP3/TGFβ signaling. Inhal Toxicol 2024; 36:217-227. [PMID: 38713814 DOI: 10.1080/08958378.2024.2335193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/15/2024] [Indexed: 05/09/2024]
Abstract
OBJECTIVE The present work concentrated on validating whether sinomenine alleviates bleomycin (BLM)-induced pulmonary fibrosis, inflammation, and oxidative stress. METHODS A rat model of pulmonary fibrosis was constructed through intratracheal injection with 5 mg/kg BLM, and the effects of 30 mg/kg sinomenine on pulmonary inflammation, fibrosis, apoptosis, and 4-hydroxynonenal density were evaluated by hematoxylin and eosin staining, Masson's trichrome staining, TUNEL staining, and immunohistochemistry. Hydroxyproline content and concentrations of inflammatory cytokines and oxidative stress markers were detected using corresponding kits. MRC-5 cells were treated with 10 ng/ml PDGF, and the effects of 1 mM sinomenine on cell proliferation were assessed by EdU assays. The mRNA expression of inflammatory cytokines and the protein levels of collagens, fibrosis markers, and key markers involved in the TLR4/NLRP3/TGFβ signaling were tested with RT-qPCR and immunoblotting analysis. RESULTS Sinomenine attenuated pulmonary fibrosis and inflammation while reducing hydroxyproline content and the protein expression of collagens and fibrosis markers in BLM-induced pulmonary fibrosis rats. Sinomenine reduced apoptosis in lung samples of BLM-challenged rats by increasing Bcl-2 and reducing Bax and cleaved caspase-3 protein expression. In addition, sinomenine alleviated inflammatory response and oxidative stress in rats with pulmonary fibrosis induced by BLM. Moreover, sinomenine inhibited the TLR4/NLRP3/TGFβ signaling pathway in lung tissues of BLM-stimulated rats. Furthermore, TLR4 inhibitor, TAK-242, attenuated PDGF-induced fibroblast proliferation and collagen synthesis in MRC-5 cells. CONCLUSION Sinomenine attenuates BLM-caused pulmonary fibrosis, inflammation, and oxidative stress by inhibiting the TLR4/NLRP3/TGFβ signaling, indicating that sinomenine might become a therapeutic candidate to treat pulmonary fibrosis.
Collapse
Affiliation(s)
- Yijue Liu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Hong Chen
- School of Medicine, Jianghan University, Wuhan city, Hubei Province, P.R. China
| | - Yan Wu
- School of Medicine, Jianghan University, Wuhan city, Hubei Province, P.R. China
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Wei Li
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Huan Peng
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Feng Gui
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Bo Yu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| |
Collapse
|
4
|
Fang K, Ren S, Zhang Q. Identification and characterization of the metabolites of sinomenine using liquid chromatography combined with benchtop Orbitrap mass spectrometry and nuclear magnetic resonance spectroscopy. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9669. [PMID: 38211350 DOI: 10.1002/rcm.9669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 01/13/2024]
Abstract
RATIONALE Sinomenine, a major bioactive compound isolated from Sinomenium acutum, has been used for the treatment of rheumatoid arthritis and other cardio-cerebrovacular diseases. However, the metabolism of this drug has not been fully investigated. The current work was carried out to investigate the in vitro metabolism of sinomenine in liver microsomes. METHODS The metabolites were generated by incubating sinomenine (3 μM) with the liver microsomes in the presence of NADPH at 37°C. The structure of the metabolites was characterized using liquid chromatography coupled to high-resolution mass spectrometry (HRMS). Two major metabolites synthesized and their structures were further confirmed using nuclear magnetic resonance spectroscopy. RESULTS Under the current conditions, 12 metabolites were found and structurally identified using high resolution MS and MS2 spectra. Among these metabolites, M1, M2, M3, M4, M5, M6, M7, M9, M11, and M12 were first reported. The metabolites M8 and M10 were synthesized and unambiguously identified as N-desmethyl-sinomenine and sinomenine N-oxide, respectively. The phenotyping study revealed that the formation of M8 was catalyzed by CYP2C8, 2C19, 2D6, and 3A4, whereas the formation of M3, M6, and M10 were exclusively catalyzed by CYP3A4. The metabolic pathways of sinomenine include N-demethylation, O-demethylation, dehydrogenation, oxygenation, and N-oxygenation. CONCLUSIONS N-Demethylation and N-oxygenation were the primary metabolic pathways of sinomenine. This study provides new insight into the in vitro metabolism of sinomenine, which would help prospects of sinomenine disposition and safety assessments.
Collapse
Affiliation(s)
- Ke Fang
- Department of Cardiology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shaoyu Ren
- Department of Cardiology, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Qian Zhang
- Department of Radiology, Shandong Provincial Third Hospital, Jinan, Shandong, China
| |
Collapse
|
5
|
Hou W, Huang L, Huang H, Liu S, Dai W, Tang J, Chen X, Lu X, Zheng Q, Zhou Z, Zhang Z, Lan J. Bioactivities and Mechanisms of Action of Sinomenine and Its Derivatives: A Comprehensive Review. Molecules 2024; 29:540. [PMID: 38276618 PMCID: PMC10818773 DOI: 10.3390/molecules29020540] [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: 12/14/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Sinomenine, an isoquinoline alkaloid extracted from the roots and stems of Sinomenium acutum, has been extensively studied for its derivatives as bioactive agents. This review concentrates on the research advancements in the biological activities and action mechanisms of sinomenine-related compounds until November 2023. The findings indicate a broad spectrum of pharmacological effects, including antitumor, anti-inflammation, neuroprotection, and immunosuppressive properties. These compounds are notably effective against breast, lung, liver, and prostate cancers, exhibiting IC50 values of approximately 121.4 nM against PC-3 and DU-145 cells, primarily through the PI3K/Akt/mTOR, NF-κB, MAPK, and JAK/STAT signaling pathways. Additionally, they manifest anti-inflammatory and analgesic effects predominantly via the NF-κB, MAPK, and Nrf2 signaling pathways. Utilized in treating rheumatic arthritis, these alkaloids also play a significant role in cardiovascular and cerebrovascular protection, as well as organ protection through the NF-κB, Nrf2, MAPK, and PI3K/Akt/mTOR signaling pathways. This review concludes with perspectives and insights on this topic, highlighting the potential of sinomenine-related compounds in clinical applications and the development of medications derived from natural products.
Collapse
Affiliation(s)
- Wen Hou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Lejun Huang
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China;
| | - Hao Huang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Shenglan Liu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Wei Dai
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jianhong Tang
- Laboratory Animal Engineering Research Center of Ganzhou, Gannan Medical University, Ganzhou 341000, China;
| | - Xiangzhao Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Xiaolu Lu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Qisheng Zheng
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Zhinuo Zhou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Ziyun Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jinxia Lan
- College of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
6
|
Abdellatif SY, Fares NH, Elsharkawy SH, Mahmoud YI. Calanus oil attenuates isoproterenol-induced cardiac hypertrophy by regulating myocardial remodeling and oxidative stress. Ultrastruct Pathol 2023; 47:12-21. [PMID: 36588172 DOI: 10.1080/01913123.2022.2163016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Calanus oil, an oil extracted from the marine crustacean Calanus finmarchicus, is one of the richest sources of omega-3 and poly-unsaturated fatty acids. Although calanus oil has been shown to have a significant anti-hypertensive, anti-inflammatory, anti-fibrotic and anti-obesity effects in various cardiovascular diseases, but little is known about its effect on pathological cardiac hypertrophy. Thus, the present study was carried out to evaluate the therapeutic effect of calanus oil on cardiac hypertrophy. Cardiac hypertrophy was induced by subcutaneous injections with isoproterenol (5 mg/kg b.w) for 14 consecutive days. Calanus oil (400 mg/kg) was given orally for 4 weeks. Cardiac pathological remodeling was evaluated by echocardiography, after which morphometric, biochemical, histological and ultrastructural analyses were performed. Calanus oil treatment significantly ameliorated isoproterenol-induced structural and functional alterations in echocardiography. Calanus oil also reduced the relative heart weight, significantly decreased the elevated cardiac enzymes (LDH and CK-MB) and the lipid peroxidation marker (MDA), augmented the myocardial antioxidant status (TAC), and ameliorated the histopathological and ultrastructural changes in cardiac tissues and prevented interstitial collagen deposition. The present study, for the first time, provided morphometric, biochemical, histological and ultrastructural evidences supporting the promising anti-hypertrophic effect of calanus oil against ISO-induced cardiac hypertrophy. This anti-hypertrophic effect of calanus oil is via regulating myocardial remodeling and oxidative stress. Therefore, it could be used as potential pharmacological intervention in the management of cardiac hypertrophy.
Collapse
Affiliation(s)
| | - Nagui H Fares
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Samar H Elsharkawy
- Department of Surgery, Anaesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Yomna I Mahmoud
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
7
|
Corynoline protects ang II-induced hypertensive heart failure by increasing PPARα and Inhibiting NF-κB pathway. Biomed Pharmacother 2022; 150:113075. [PMID: 35658238 DOI: 10.1016/j.biopha.2022.113075] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Heart failure is a fairly common outcome of hypertension. Recent studies have highlighted the key role of the non-hemodynamic activity of angiotensin II (Ang II) in hypertensive heart failure via inducing cardiac inflammation. Drugs that disrupt Ang II-induced cardiac inflammation may have clinical utility in the treatment of hypertensive heart failure. A naturally occurring compound, corynoline, exhibit anti-inflammatory activities in other systems. C57BL/6 mice were injected with Ang II via a micro-osmotic pump for four weeks to develop hypertensive heart failure. The mice were treated with corynoline by gavage for two weeks. RNA-sequencing analysis was performed to explore the potential mechanism of corynoline. We found that corynoline could inhibit inflammation, myocardial fibrosis, and hypertrophy to prevent heart dysfunction, without the alteration of blood pressure. RNA-sequencing analysis indicates that the PPARα pathway is involved Ang II-induced cardiac fibrosis and cardiac remodeling. Corynoline reversed Ang II-induced PPARα inhibition both in vitro and in vivo. We further found that corynoline increases the interaction between PPARα and P65 to inhibit the NF-κB pro-inflammatory pathway in H9c2 cells. Our studies show that corynoline relieves Ang II-induced hypertensive heart failure by increasing the interaction between PPARα and P65 to inhibit the NF-κB pathway.
Collapse
|
8
|
Lu C, Guo X, He X, Chang Y, Zheng F, Xu C, Zhang S, Zhou Y, Li J. Cardioprotective effects of Sinomenine in myocardial ischemia/reperfusion injury in a rat model. Saudi Pharm J 2022; 30:669-678. [PMID: 35812144 PMCID: PMC9257858 DOI: 10.1016/j.jsps.2022.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/03/2022] [Indexed: 02/08/2023] Open
Abstract
Background Ischemia reperfusion (I/R) play an imperative role in the expansion of cardiovascular disease. Sinomenine (SM) has been exhibited to possess antioxidant, anticancer, anti-inflammatory, antiviral and anticarcinogenic properties. The aim of the study was scrutinized the cardioprotective effect of SM against I/R injury in rat. Methods Rat were randomly divided into normal control (NC), I/R control and I/R + SM (5, 10 and 20 mg/kg), respectively. Ventricular arrhythmias, body weight and heart weight were estimated. Antioxidant, inflammatory cytokines, inflammatory mediators and plasmin system indicator were accessed. Results Pre-treated SM group rats exhibited the reduction in the duration and incidence of ventricular fibrillation, ventricular ectopic beat (VEB) and ventricular tachycardia along with suppression of arrhythmia score during the ischemia (30 and 120 min). SM treated rats significantly (P < 0.001) altered the level of antioxidant parameters. SM treatment significantly (P < 0.001) repressed the level of creatine kinase MB (CK-MB), creatine kinase (CK) and troponin I (Tnl). SM treated rats significantly (P < 0.001) repressed the tissue factor (TF), thromboxane B2 (TXB2), plasminogen activator inhibitor 1 (PAI-1) and plasma fibrinogen (Fbg) and inflammatory cytokines and inflammatory mediators. Conclusion Our result clearly indicated that SM plays anti-arrhythmia effect in I/R injury in the rats via alteration of oxidative stress and inflammatory reaction.
Collapse
|
9
|
Yuan M, Zhao B, Jia H, Zhang C, Zuo X. Sinomenine ameliorates cardiac hypertrophy by activating Nrf2/ARE signaling pathway. Bioengineered 2021; 12:12778-12788. [PMID: 34895050 PMCID: PMC8810090 DOI: 10.1080/21655979.2021.2000195] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 02/04/2023] Open
Abstract
Cardiac hypertrophy (CH) is a result of the physiological adaptation of the heart to coronary heart disease, hypertension, and other cardiovascular diseases. Sinomenine is extracted from Caulis Sinomenii. This study aimed to explore the specific mechanism of the action of sinomenine in cardiac hypertrophy (CH) via Nrf2/ARE signaling pathway in vivo and in vitro. To establish a model of CH, H9C2 cells were treated with angiotensin II (Ang II) and intraperitoneally injected with isoproterenol. Then the cells were treated with 50 and 100 μM sinomenine. TUNEL, HE, rhodamine-labeled phalloidin, and immunohistochemical staining were performed. Flow cytometry was used to measure apoptosis rates. mRNA expression of ANP, BNP, and β-MHC was determined by qRT-PCR. Furthermore, western blotting was performed to analyze protein expression. After sinomenine treatment, the surface area and apoptosis rates were decreased. Furthermore, the mRNA expression of ANP, BNP, and β-MHC, levels of reactive oxygen species and malondialdehyde, and protein expression of Caspase3 and Bax were down-regulated, and the protein expression of Bcl-2 was upregulated. Sinomenine activates the Nrf2/ARE signaling pathway, and inhibition of this signaling pathway reversed the effects of sinomenine. In animal experiments, sinomenine decreased the heart weight and left ventricular weight indices, as well as the expression of ANP, BNP, and β-MHC. Furthermore, sinomenine reduced the apoptosis rate and relieved CH-related oxidative stress by activating the Nrf2/ARE signaling pathway. Together, these findings reveal that sinomenine is a potential candidate drug for CH treatment and further research is required to generalize the result in human subjects.
Collapse
Affiliation(s)
- ManLi Yuan
- Department of Ultrasound Medicine, PLA Strategic Support Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Bei Zhao
- Department of Cardiovascular Medicine, PLA Strategic Support Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Huaping Jia
- Department of Ultrasound Medicine, PLA Strategic Support Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Can Zhang
- Department of Ultrasound Medicine, PLA Strategic Support Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Xiaowen Zuo
- Department of Ultrasound Medicine, PLA Strategic Support Force Characteristic Medical Center, Beijing, People's Republic of China
| |
Collapse
|