1
|
Song C, Huang L, Li D, Zhao X. Artemisinin Alleviates Alcohol-Induced Cardiotoxicity by Inhibiting Ferroptosis via the Nrf2/NQO1 Pathway In Vivo and In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12940-12948. [PMID: 40384105 DOI: 10.1021/acs.jafc.4c08388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
The present study was designed to explore the protective effects of artemisinin on alcohol-induced cardiac injuries and its mechanisms. In H9c2 cells, cell viability, reactive oxygen species (ROS), labile iron pool (LIP), and mitochondrial membrane potential (MMP) were measured. In the mouse model of alcohol-induced cardiomyopathy, body weight and electrocardiogram (ECG) were recorded every day. Heart tissue creatine kinase (CK), lactic dehydrogenase (LDH), iron, glutathione (GSH), malondialdehyde (MDA), and histological examination were measured. Western blot assay was performed to evaluate the expression of ferroptosis-related proteins in vitro and in vivo. The results in vitro showed that cell viability was increased, ROS and LIP contents were decreased, and the level of MMP was increased in artemisinin-treated H9c2 cells. Tissues CK, LDH, and GSH were improved after being treated with artemisinin. The ferroptosis biomarkers, including MDA and tissue iron, were attenuated after artemisinin treatment. Artemisinin protected the heart from alcohol damage by ECG and histological examination. Additionally, artemisinin down-regulated the expression of TfR and P53 and up-regulated Nrf2, HO-1, NQO1, and GPX4 expressions in vitro and in vivo. The results showed that both Fer-1 and artemisinin abolished ferroptosis. The data presented here showed that artemisinin had the potential to protect alcohol-induced cardiotoxicity through the inhibition of Nrf2/NQO1-dependent ferroptosis.
Collapse
Affiliation(s)
- Chunpu Song
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Ling Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Dongjie Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiaoyan Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Engineering Research Center of Coptis Development & Utilization, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
2
|
Jeong HK, Yoon N, Kim YR, Lee KH, Park HW. Artemisinin-Quinidine Combination for Suppressing Ventricular Tachyarrhythmia in an Ex Vivo Model of Brugada Syndrome. J Korean Med Sci 2025; 40:e2. [PMID: 39763307 PMCID: PMC11707659 DOI: 10.3346/jkms.2025.40.e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/12/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The ionic mechanism underlying Brugada syndrome (BrS) arises from an imbalance in transient outward current flow between the epicardium and endocardium. Previous studies report that artemisinin, originally derived from a Chinese herb for antimalarial use, inhibits the Ito current in canines. In a prior study, we showed the antiarrhythmic effects of artemisinin in BrS wedge preparation models. However, quinidine remains a well-established antiarrhythmic agent for treating BrS. Therefore, this study aims to investigate the efficacy of combining artemisinin with low-dose quinidine in suppressing ventricular tachyarrhythmia (VTA) in experimental canine BrS models. METHODS Transmural pseudo-electrocardiogram and epicardial/endocardial action potential (AP) were recorded from coronary-perfused canine right ventricular wedge preparation. To mimic the BrS model, acetylcholine (3 μM), calcium channel blocker verapamil (1 μM), and Ito agonist NS5806 (6-10 μM) were administered until VTA was induced. Subsequently, low-dose quinidine (1-2 μM) combined with artemisinin (100 μM) was perfused to mitigate VTA. Key parameters, including AP duration, J wave area, notch index, and T wave dispersion, were measured. RESULTS After administering the provocation agents, all sample models exhibited prominent J waves and VTA. Artemisinin alone (100-150 μM) suppressed VTA and restored the AP dome in all three preparations. Its infusion resulted in reductions in the J wave area and epicardial notch index. Consequently, low-dose quinidine (1-2 μM) did not fully restore the AP dome in all six sample models. However, when combined with additional artemisinin (100 μM), low-dose quinidine effectively suppressed VTA in all six models and restored the AP dome while also decreasing the J wave area and epicardial notch index. CONCLUSION Low-dose quinidine alone fails to fully alleviate VTA in the BrS wedge model. However, its combination with artemisinin effectively suppresses VTA. Artemisinin may reduce the therapeutic dose of quinidine, potentially minimizing its associated adverse effects.
Collapse
Affiliation(s)
- Hyung Ki Jeong
- Division of Cardiology, Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Korea
- Institute of Wonkang Medical Science, Iksan, Korea
| | - Namsik Yoon
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea.
| | - Yoo Ri Kim
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Ki Hong Lee
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Hyung Wook Park
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
3
|
Afshari H, Noori S, Zarghi A. Curcumin potentiates the anti-inflammatory effects of Tehranolide by modulating the STAT3/NF-κB signaling pathway in breast and ovarian cancer cell lines. Inflammopharmacology 2023; 31:2541-2555. [PMID: 37452228 DOI: 10.1007/s10787-023-01281-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Studies have demonstrated that natural products, such as curcumin and artemisinin, possess anti-inflammatory effects, which can be beneficial for cancer treatment. Tehranolide, as a novel natural product, has a wide range of biological activities, including anti-cancer effects. However, many properties of Tehranolide, like its anti-inflammatory activity and its combination with curcumin, have not been investigated yet. This investigation examined the anti-inflammatory activity of Tehranolide, either alone or in combination with curcumin, via modulating the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and STAT3 (signal transducer and activator of transcription 3) signaling pathways in MDA-MB-231 and SKOV3, breast and ovarian cancer cell lines. METHODS ELISA-based methods were employed to measure the pro-inflammatory cytokine levels and the NF-κB activity in lipopolysaccharide (LPS)-induced cells. The real-time PCR experiment and Griess test were performed to evaluate inducible nitric oxide synthase (iNOS) gene expression and nitrite levels, respectively. The STAT3 and NF-κB signaling pathways were investigated by Western blotting analysis. Tehranolide's anti-cancer activity was also assessed in a mouse model of breast cancer using the TUNEL (terminal deoxynucleotidyl transferase nick-end labeling) assay. RESULTS Tehranolide diminished levels of pro-inflammatory cytokines in cancer cells. Additionally, it suppressed NF-κB DNA binding and STAT3 phosphorylation, reducing iNOS gene expression and nitrite production. Moreover, Western blotting showed that Tehranolide enhanced the inhibitory κB (IκBα) and Bcl-2 (B-cell lymphoma 2)-associated X (BAX) expression, and downregulated the expression of Bcl-2 proteins. Furthermore, the TUNEL assay demonstrated that Tehranolide induced apoptosis in a breast cancer mouse model. Curcumin potentiated all the anti-inflammatory effects of Tehranolide. CONCLUSION This investigation indicated for the first time that Tehranolide, either alone or in combination with curcumin, exerted its anti-inflammatory effects by suppressing NF-κB and STAT3 signaling pathways in SKOV3 and MDA-MB-231 cells.
Collapse
Affiliation(s)
- Havva Afshari
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokoofe Noori
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Farrag EAE, Hammad MO, Safwat SM, Hamed S, Hellal D. Artemisinin attenuates type 2 diabetic cardiomyopathy in rats through modulation of AGE-RAGE/HMGB-1 signaling pathway. Sci Rep 2023; 13:11043. [PMID: 37422477 PMCID: PMC10329689 DOI: 10.1038/s41598-023-37678-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/26/2023] [Indexed: 07/10/2023] Open
Abstract
Diabetes mellitus is a common metabolic disorder. About two-thirds of diabetic patients develop diabetic cardiomyopathy (DCM), which becomes a challenging issue as it severely threatens the patient's life. Hyperglycemia and the resulting advanced glycated end products (AGE) and their receptor (RAGE)/High Mobility Group Box-1 (HMGB-1) molecular pathway are thought to be key players. Recently, artemisinin (ART) has gained more attention owing to its potent biological activities beyond its antimalarial effect. Herein, we aim to evaluate the effect of ART on DCM and the possible underlying mechanisms. Twenty-four male Sprague-Dawley rats were divided into: control, ART, type 2 diabetic and type 2 diabetic treated with ART groups. At the end of the research, the ECG was recorded, then the heart weight to body weight (HW/BW) ratio, fasting blood glucose, serum insulin and HOMA-IR were evaluated. Cardiac biomarkers (CK-MB and LDH), oxidative stress markers, IL-1β, AGE, RAGE and HMGB-1 expression were also measured. The heart specimens were stained for H&E as well as Masson's trichrome. DCM induced disturbances in all studied parameters; contrary to this, ART improved these insults. Our study concluded that ART could improve DCM through modulation of the AGE-RAGE/HMGB-1 signaling pathway, with subsequent impacts on oxidative stress, inflammation and fibrosis. ART could therefore be a promising therapy for the management of DCM.
Collapse
Affiliation(s)
- Eman A E Farrag
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Maha O Hammad
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Sally M Safwat
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shereen Hamed
- Department of Medical Histology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Hellal
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
5
|
Posadino AM, Giordo R, Pintus G, Mohammed SA, Orhan IE, Fokou PVT, Sharopov F, Adetunji CO, Gulsunoglu-Konuskan Z, Ydyrys A, Armstrong L, Sytar O, Martorell M, Razis AFA, Modu B, Calina D, Habtemariam S, Sharifi-Rad J, Cho WC. Medicinal and mechanistic overview of artemisinin in the treatment of human diseases. Biomed Pharmacother 2023; 163:114866. [PMID: 37182516 DOI: 10.1016/j.biopha.2023.114866] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023] Open
Abstract
Artemisinin (ART) is a bioactive compound isolated from the plant Artemisia annua and has been traditionally used to treat conditions such as malaria, cancer, viral infections, bacterial infections, and some cardiovascular diseases, especially in Asia, North America, Europe and other parts of the world. This comprehensive review aims to update the biomedical potential of ART and its derivatives for treating human diseases highlighting its pharmacokinetic and pharmacological properties based on the results of experimental pharmacological studies in vitro and in vivo. Cellular and molecular mechanisms of action, tested doses and toxic effects of artemisinin were also described. The analysis of data based on an up-to-date literature search showed that ART and its derivatives display anticancer effects along with a wide range of pharmacological activities such as antibacterial, antiviral, antimalarial, antioxidant and cardioprotective effects. These compounds have great potential for discovering new drugs used as adjunctive therapies in cancer and various other diseases. Detailed translational and experimental studies are however needed to fully understand the pharmacological effects of these compounds.
Collapse
Affiliation(s)
- Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Roberta Giordo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy; Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah 27272, United Arab Emirates
| | - Soheb Anwar Mohammed
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, University of Pittsburgh, PA 15213, USA
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey; Turkish Academy of Sciences (TÜBA), Vedat Dalokay Cad., No. 112, 06670 Ankara, Turkey
| | | | - Farukh Sharopov
- V.I. Nikitin Chemistry Institute of the National Academy of Sciences of Tajikistan, Ayni 299/2, 734063 Dushanbe, Tajikistan
| | - Charles Oluwaseun Adetunji
- Applied Microbiology, Biotechnology and Nanotechnology Laboratory, Department of Microbiology, Edo State University Uzairue, Iyamho, PMB 04 Auchi, Edo State, Nigeria
| | - Zehra Gulsunoglu-Konuskan
- Faculty of Health Science, Nutrition and Dietetics Department, Istanbul Aydin University, Istanbul 34295, Turkey
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi ave. 71, 050040 Almaty, Kazakhstan
| | - Lorene Armstrong
- State University of Ponta Grossa, Departament of Pharmaceutical Sciences, 84030900 Ponta Grossa, Paraná, Brazil; Federal University of Paraná, Department of Pharmacy, 80210170 Curitiba, Paraná, Brazil
| | - Oksana Sytar
- Institute of Plant and Environmental Sciences, Slovak Agricultural University in Nitra, 94976 Nitra, Slovakia
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, 4070386 Concepción, Chile; Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, 4070386 Concepción, Chile.
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Babagana Modu
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Department of Biochemistry, Faculty of Science, University of Maiduguri, 1069 Maiduguri, Borno State, Nigeria
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| | | | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
6
|
Shinyuy LM, Loe GE, Jansen O, Mamede L, Ledoux A, Noukimi SF, Abenwie SN, Ghogomu SM, Souopgui J, Robert A, Demeyer K, Frederich M. Secondary Metabolites Isolated from Artemisia afra and Artemisia annua and Their Anti-Malarial, Anti-Inflammatory and Immunomodulating Properties-Pharmacokinetics and Pharmacodynamics: A Review. Metabolites 2023; 13:metabo13050613. [PMID: 37233654 DOI: 10.3390/metabo13050613] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
There are over 500 species of the genus Artemisia in the Asteraceae family distributed over the globe, with varying potentials to treat different ailments. Following the isolation of artemisinin (a potent anti-malarial compound with a sesquiterpene backbone) from Artemisia annua, the phytochemical composition of this species has been of interest over recent decades. Additionally, the number of phytochemical investigations of other species, including those of Artemisia afra in a search for new molecules with pharmacological potentials, has increased in recent years. This has led to the isolation of several compounds from both species, including a majority of monoterpenes, sesquiterpenes, and polyphenols with varying pharmacological activities. This review aims to discuss the most important compounds present in both plant species with anti-malarial properties, anti-inflammatory potentials, and immunomodulating properties, with an emphasis on their pharmacokinetics and pharmacodynamics properties. Additionally, the toxicity of both plants and their anti-malaria properties, including those of other species in the genus Artemisia, is discussed. As such, data were collected via a thorough literature search in web databases, such as ResearchGate, ScienceDirect, Google scholar, PubMed, Phytochemical and Ethnobotanical databases, up to 2022. A distinction was made between compounds involved in a direct anti-plasmodial activity and those expressing anti-inflammatory and immunomodulating activities or anti-fever properties. For pharmacokinetics activities, a distinction was made between compounds influencing bioavailability (CYP effect or P-Glycoprotein effect) and those affecting the stability of pharmacodynamic active components.
Collapse
Affiliation(s)
- Lahngong Methodius Shinyuy
- Laboratory of Pharmacognosy, Department of Pharmacy, Center of Interdisciplinary Research on Medicine (CIRM), University of Liege, 4000 Liège, Belgium
- Laboratory of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Department of Analytical, Applied Chemometrics and Molecular Modeling (FABI), Faculty of Medicine and Pharmacy, Vrije Universiteit of Brussel, 1050 Ixelles, Belgium
- Laboratory of Pharmacochemical and Natural Pharmaceutical Substances, Doctoral Training Unit in Health Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala P.O. Box 2701, Cameroon
| | - Gisèle E Loe
- Laboratory of Pharmacochemical and Natural Pharmaceutical Substances, Doctoral Training Unit in Health Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala P.O. Box 2701, Cameroon
| | - Olivia Jansen
- Laboratory of Pharmacognosy, Department of Pharmacy, Center of Interdisciplinary Research on Medicine (CIRM), University of Liege, 4000 Liège, Belgium
| | - Lúcia Mamede
- Laboratory of Pharmacognosy, Department of Pharmacy, Center of Interdisciplinary Research on Medicine (CIRM), University of Liege, 4000 Liège, Belgium
| | - Allison Ledoux
- Laboratory of Pharmacognosy, Department of Pharmacy, Center of Interdisciplinary Research on Medicine (CIRM), University of Liege, 4000 Liège, Belgium
| | - Sandra Fankem Noukimi
- Molecular and Cell Biology Laboratory (MCBL), Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon
- Embryology and Biotechnology Laboratory, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Suh Nchang Abenwie
- Epidemiology and Biostatistics Unit (EPiD), Institute of Clinical and Experimental Research (IREC), UCLouvain, 1200 Brussel, Belgium
| | - Stephen Mbigha Ghogomu
- Molecular and Cell Biology Laboratory (MCBL), Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon
| | - Jacob Souopgui
- Embryology and Biotechnology Laboratory, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Annie Robert
- Epidemiology and Biostatistics Unit (EPiD), Institute of Clinical and Experimental Research (IREC), UCLouvain, 1200 Brussel, Belgium
| | - Kristiaan Demeyer
- Laboratory of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Department of Analytical, Applied Chemometrics and Molecular Modeling (FABI), Faculty of Medicine and Pharmacy, Vrije Universiteit of Brussel, 1050 Ixelles, Belgium
| | - Michel Frederich
- Laboratory of Pharmacognosy, Department of Pharmacy, Center of Interdisciplinary Research on Medicine (CIRM), University of Liege, 4000 Liège, Belgium
| |
Collapse
|
7
|
Artemisinin and Its Derivate Alleviate Pulmonary Hypertension and Vasoconstriction in Rodent Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2782429. [PMID: 35757500 PMCID: PMC9232380 DOI: 10.1155/2022/2782429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 03/20/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022]
Abstract
Background Pulmonary arterial hypertension (PAH) is a complex pulmonary vasculature disease characterized by progressive obliteration of small pulmonary arteries and persistent increase in pulmonary vascular resistance, resulting in right heart failure and death if left untreated. Artemisinin (ARS) and its derivatives, which are common antimalarial drugs, have been found to possess a broad range of biological effects. Here, we sought to determine the therapeutic benefit and mechanism of ARS and its derivatives treatment in experimental pulmonary hypertension (PH) models. Methods Isolated perfused/ventilated lung and isometric tension measurements in arteries were performed to test pulmonary vasoconstriction and relaxation. Monocrotaline (MCT) and hypoxia+Su5416 (SuHx) were administered to rats to induce severe PH. Evaluation methods of ARS treatment and its derivatives in animal models include echocardiography, hemodynamics measurement, and histological staining. In vitro, the effect of these drugs on proliferation, viability, and hypoxia-inducible factor 1α (HIF1α) was examined in human pulmonary arterial smooth muscle cells (hPASMCs). Results ARS treatment attenuated pulmonary vasoconstriction induced by high K+ solution or alveolar hypoxia, decreased pulmonary artery (PA) basal vascular tension, improved acetylcholine- (ACh-) induced endothelial-dependent relaxation, increased endothelial nitric oxide (NO) synthase (eNOS) activity and NO levels, and decreased levels of NAD(P)H oxidase subunits (NOX2 and NOX4) expression, NAD(P)H oxidase activity, and reactive oxygen species (ROS) levels of pulmonary arteries (PAs) in MCT-PH rats. NOS inhibitor, L-NAME, abrogated the effects of ARS on PA constriction and relaxation. Furthermore, chronic application of both ARS and its derivative dihydroartemisinin (DHA) attenuated right ventricular systolic pressure (RVSP), Fulton index (right ventricular hypertrophy), and vascular remodeling of PAs in the two rat PH models. In addition, DHA inhibited proliferation and migration of hypoxia-induced PASMCs. Conclusions In conclusion, these results indicate that treatment with ARS or DHA can inhibit PA vasoconstriction, PASMC proliferation and migration, and vascular remodeling, as well as improve PA endothelium-dependent relaxation, and eventually attenuate the development and progression of PH. These effects might be achieved by decreasing NAD(P)H oxidase generated ROS production and increasing eNOS activation to release NO in PAs. ARS and its derivatives might have the potential to be novel drugs for the treatment of PH.
Collapse
|
8
|
Farmanpour-Kalalagh K, Beyraghdar Kashkooli A, Babaei A, Rezaei A, van der Krol AR. Artemisinins in Combating Viral Infections Like SARS-CoV-2, Inflammation and Cancers and Options to Meet Increased Global Demand. FRONTIERS IN PLANT SCIENCE 2022; 13:780257. [PMID: 35197994 PMCID: PMC8859114 DOI: 10.3389/fpls.2022.780257] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/03/2022] [Indexed: 05/05/2023]
Abstract
Artemisinin is a natural bioactive sesquiterpene lactone containing an unusual endoperoxide 1, 2, 4-trioxane ring. It is derived from the herbal medicinal plant Artemisia annua and is best known for its use in treatment of malaria. However, recent studies also indicate the potential for artemisinin and related compounds, commonly referred to as artemisinins, in combating viral infections, inflammation and certain cancers. Moreover, the different potential modes of action of artemisinins make these compounds also potentially relevant to the challenges the world faces in the COVID-19 pandemic. Initial studies indicate positive effects of artemisinin or Artemisia spp. extracts to combat SARS-CoV-2 infection or COVID-19 related symptoms and WHO-supervised clinical studies on the potential of artemisinins to combat COVID-19 are now in progress. However, implementing multiple potential new uses of artemisinins will require effective solutions to boost production, either by enhancing synthesis in A. annua itself or through biotechnological engineering in alternative biosynthesis platforms. Because of this renewed interest in artemisinin and its derivatives, here we review its modes of action, its potential application in different diseases including COVID-19, its biosynthesis and future options to boost production.
Collapse
Affiliation(s)
- Karim Farmanpour-Kalalagh
- Department of Horticultural Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Arman Beyraghdar Kashkooli
- Department of Horticultural Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
- *Correspondence: Arman Beyraghdar Kashkooli,
| | - Alireza Babaei
- Department of Horticultural Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Ali Rezaei
- Department of Horticultural Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
9
|
Liu X, Wang X, Pan Y, Zhao L, Sun S, Luo A, Bao C, Tang H, Han Y. Artemisinin Improves Acetylcholine-Induced Vasodilatation in Rats with Primary Hypertension. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:4489-4502. [PMID: 34764635 PMCID: PMC8576437 DOI: 10.2147/dddt.s330721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022]
Abstract
Purpose Endothelial dysfunction and the subsequent decrease in endothelium-dependent vascular relaxation of small arteries are major features of hypertension. Artemisinin, a well-known antimalarial drug, has been shown to exert protecting roles against endothelial cell injury in cardiac and pulmonary vascular diseases. The current study aimed to investigate the effects of artemisinin on endothelium-dependent vascular relaxation and arterial blood pressure, as well as the potential signalling pathways in spontaneously hypertensive rats (SHRs). Methods In this study, acetylcholine (ACh)-induced dose-dependent relaxation assays were performed to evaluate vascular endothelial function after treatment with artemisinin. Artemisinin was administered to the rats by intravenous injection or to arteries by incubation for the acute exposure experiments, and it was administered to rats by intraperitoneal injection for 28 days for the chronic experiments. Results Both acute and chronic administration of artemisinin decreased the heart rate and improved ACh-induced endothelium-dependent relaxation but negligibly affected the arterial blood pressure in SHRs. Incubation with artemisinin decreased basal vascular tension, NAD(P)H oxidase activity and reactive oxygen species (ROS) levels, but it also increased endothelial nitric oxide (NO) synthase (eNOS) activity and NO levels in the mesenteric artery, coronary artery, and pulmonary artery of SHRs. Artemisinin chronic administration to SHRs increased the protein expression of eNOS and decreased the protein expression of the NAD(P)H oxidase subunits NOX-2 and NOX-4 in the mesenteric artery. Conclusion These results indicate that treatment with artemisinin has beneficial effects on reducing the heart rate and basal vascular tension and improving endothelium-dependent vascular relaxation in hypertension, which might occur by increasing eNOS activation and NO release and inhibiting NAD(P)H oxidase derived ROS production.
Collapse
Affiliation(s)
- Xuanxuan Liu
- Key Laboratory of Targeted Intervention for Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,Department of Physiology and Pathologic Physiology, Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, People's Republic of China
| | - Xingxing Wang
- Key Laboratory of Targeted Intervention for Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yan Pan
- Key Laboratory of Targeted Intervention for Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Li Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Shuo Sun
- Key Laboratory of Targeted Intervention for Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ang Luo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Changlei Bao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ying Han
- Key Laboratory of Targeted Intervention for Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
10
|
Phytochemicals as potential IKK-β inhibitor for the treatment of cardiovascular diseases in plant preservation: terpenoids, alkaloids, and quinones. Inflammopharmacology 2019; 28:83-93. [DOI: 10.1007/s10787-019-00640-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
|
11
|
Yin J, Xia W, Zhang Y, Ding G, Chen L, Yang G, Huang S, Jia Z, Zhang A. Role of dihydroartemisinin in regulating prostaglandin E2 synthesis cascade and inflammation in endothelial cells. Heart Vessels 2018; 33:1411-1422. [DOI: 10.1007/s00380-018-1190-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 05/18/2018] [Indexed: 10/16/2022]
|
12
|
Seo EJ, Fischer N, Efferth T. Phytochemicals as inhibitors of NF-κB for treatment of Alzheimer’s disease. Pharmacol Res 2018; 129:262-273. [DOI: 10.1016/j.phrs.2017.11.030] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/19/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022]
|
13
|
Zhao L, Wu D, Sang M, Xu Y, Liu Z, Wu Q. Stachydrine ameliorates isoproterenol-induced cardiac hypertrophy and fibrosis by suppressing inflammation and oxidative stress through inhibiting NF-κB and JAK/STAT signaling pathways in rats. Int Immunopharmacol 2017; 48:102-109. [PMID: 28499193 DOI: 10.1016/j.intimp.2017.05.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/21/2017] [Accepted: 05/03/2017] [Indexed: 12/18/2022]
Abstract
Cardiac hypertrophy (CH), as one of the major causes of morbidity and mortality in the world, has become an independent and predictive risk factor for adverse cardiovascular events. However, progress in treatment remains sluggish in recent years. Therefore, compounds derived from non-toxic nature plants are urgently needed. Stachydrine (STA), which is isolated from Leonurus, has various activities, including resistance to cardiovascular disease, but little is known about its effect on CH or the mechanisms. We herein investigated the effect of STA on isoproterenol-induced CH and the underlying mechanisms. Treatment with STA significantly increased the ratios of heart weight/body weight, left ventricle weight/body weight and the cross-sectional areas of cardiomyocytes. In addition, STA significantly decreased the mRNA levels of atrial natriuretic peptide, B-type natriuretic peptide and β-myosin heavy chain. Furthermore, isoproterenol-induced fibrosis in rats receiving STA was significant attenuated, as evidenced by decreased ratio of fibrotic area/total area and decreased mRNA levels of collagens I and III. Given down-regulation of interleukin-6, tumor necrosis factor-α, interferon-γ (IFN-γ) and IFN-1β, treatment with STA significantly reversed the expressions of pro-inflammatory induced by isoproterenol. Moreover, STA attenuated the oxidative stress level in serum of isoproterenol-induced CH rats, as shown by increased activity of superoxide dismutase and decreased malondialdehyde level. STA inhibited the expressions of phosphorylated IκBα, NF-κB p65, JAK2 and STAT3 in vivo. Thus, both NF-κB and JAK/STAT signalings played essential roles in mediating the anti-CH effect of STA. Collectively, STA has a potent protective effect on isoproterenol-induced CH, with therapeutic implication for CH.
Collapse
Affiliation(s)
- Lingling Zhao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Dawei Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Mengru Sang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Yiming Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Zhaoguo Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Qinan Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China; Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China; National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, China.
| |
Collapse
|
14
|
Reid BG, Stratton MS, Bowers S, Cavasin MA, Demos-Davies KM, Susano I, McKinsey TA. Discovery of novel small molecule inhibitors of cardiac hypertrophy using high throughput, high content imaging. J Mol Cell Cardiol 2016; 97:106-13. [PMID: 27130278 PMCID: PMC5002372 DOI: 10.1016/j.yjmcc.2016.04.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/15/2016] [Accepted: 04/25/2016] [Indexed: 12/11/2022]
Abstract
Chronic cardiac hypertrophy is maladaptive and contributes to the pathogenesis of heart failure. The objective of this study was to identify small molecule inhibitors of pathological cardiomyocyte hypertrophy. High content screening was performed with primary neonatal rat ventricular myocytes (NRVMs) cultured on 96-well plates and treated with a library of 3241 distinct small molecules. Non-toxic hit compounds that blocked hypertrophy in response to phenylephrine (PE) and phorbol myristate acetate (PMA) were identified based on their ability to reduce cell size and inhibit expression of atrial natriuretic factor (ANF), which is a biomarker of pathological cardiac hypertrophy. Many of the hit compounds are existing drugs that have not previously been evaluated for benefit in the setting of cardiovascular disease. One such compound, the anti-malarial drug artesunate, blocked left ventricular hypertrophy (LVH) and improved cardiac function in adult mice subjected to transverse aortic constriction (TAC). These findings demonstrate that phenotypic screening with primary cardiomyocytes can be used to discover anti-hypertrophic lead compounds for heart failure drug discovery. Using annotated libraries of compounds with known selectivity profiles, this screening methodology also facilitates chemical biological dissection of signaling networks that control pathological growth of the heart.
Collapse
Affiliation(s)
- Brian G Reid
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, United States
| | - Matthew S Stratton
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States; Department of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Anschutz Medical Campus, United States
| | - Samantha Bowers
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, United States
| | - Maria A Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States; Department of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Anschutz Medical Campus, United States
| | - Kimberley M Demos-Davies
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States
| | - Isidro Susano
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, United States
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States; Department of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Anschutz Medical Campus, United States.
| |
Collapse
|
15
|
Pedraza-Alva G, Pérez-Martínez L, Valdez-Hernández L, Meza-Sosa KF, Ando-Kuri M. Negative regulation of the inflammasome: keeping inflammation under control. Immunol Rev 2016; 265:231-57. [PMID: 25879297 DOI: 10.1111/imr.12294] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In addition to its roles in controlling infection and tissue repair, inflammation plays a critical role in diverse and distinct chronic diseases, such as cancer, metabolic syndrome, and neurodegenerative disorders, underscoring the harmful effect of an uncontrolled inflammatory response. Regardless of the nature of the stimulus, initiation of the inflammatory response is mediated by assembly of a multimolecular protein complex called the inflammasome, which is responsible for the production of inflammatory cytokines, such as interleukin-1β (IL-1β) and IL-18. The different stimuli and mechanisms that control inflammasome activation are fairly well understood, but the mechanisms underlying the control of undesired inflammasome activation and its inactivation remain largely unknown. Here, we review recent advances in our understanding of the molecular mechanisms that negatively regulate inflammasome activation to prevent unwanted activation in the resting state, as well as those involved in terminating the inflammatory response after a specific insult to maintain homeostasis.
Collapse
Affiliation(s)
- Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | | | | | | | | |
Collapse
|
16
|
Artemisinin triggers a G1 cell cycle arrest of human Ishikawa endometrial cancer cells and inhibits cyclin-dependent kinase-4 promoter activity and expression by disrupting nuclear factor-κB transcriptional signaling. Anticancer Drugs 2014; 25:270-81. [PMID: 24296733 DOI: 10.1097/cad.0000000000000054] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Relatively little is known about the antiproliferative effects of artemisinin, a naturally occurring antimalarial compound from Artemisia annua, or sweet wormwood, in human endometrial cancer cells. Artemisinin induced a G1 cell cycle arrest in cultured human Ishikawa endometrial cancer cells and downregulated cyclin-dependent kinase-2 (CDK2) and CDK4 transcript and protein levels. Analysis of CDK4 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK4 gene expression was accounted for by the loss of CDK4 promoter activity. Chromatin immunoprecipitation demonstrated that artemisinin inhibited nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) subunit p65 and p50 interactions with the endogenous Ishikawa cell CDK4 promoter. Coimmunoprecipitation revealed that artemisinin disrupts endogenous p65 and p50 nuclear translocation through increased protein-protein interactions with IκB-α, an NF-κB inhibitor, and disrupts its interaction with the CDK4 promoter, leading to a loss of CDK4 gene expression. Artemisinin treatment stimulated the cellular levels of IκB-α protein without altering the level of IκB-α transcripts. Finally, expression of exogenous p65 resulted in the accumulation of this NF-κB subunit in the nucleus of artemisinin-treated and artemisinin-untreated cells, reversed the artemisinin downregulation of CDK4 protein expression and promoter activity, and prevented the artemisinin-induced G1 cell cycle arrest. Taken together, our results demonstrate that a key event in the artemisinin antiproliferative effects in endometrial cancer cells is the transcriptional downregulation of CDK4 expression by disruption of NF-κB interactions with the CDK4 promoter.
Collapse
|
17
|
GU YONGWEI, WU GANG, WANG XIN, WANG XI, WANG YULIN, HUANG CONGXIN. Artemisinin prevents electric remodeling following myocardial infarction possibly by upregulating the expression of connexin 43. Mol Med Rep 2014; 10:1851-6. [DOI: 10.3892/mmr.2014.2431] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 03/28/2014] [Indexed: 11/06/2022] Open
|
18
|
Shi JQ, Zhang CC, Sun XL, Cheng XX, Wang JB, Zhang YD, Xu J, Zou HQ. Antimalarial drug artemisinin extenuates amyloidogenesis and neuroinflammation in APPswe/PS1dE9 transgenic mice via inhibition of nuclear factor-κB and NLRP3 inflammasome activation. CNS Neurosci Ther 2013; 19:262-8. [PMID: 23406388 DOI: 10.1111/cns.12066] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 01/14/2013] [Accepted: 01/14/2013] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The activation of nuclear factor-kappa B (NF-κB) and NLRP3 inflammasome is involved in neuroinflammation, which is closely linked to Alzheimer's disease (AD). In vivo and in vitro studies have suggested that artemisinin shows antiinflammatory effects in inflammation-related diseases. However, the impacts of artemisinin on AD have not been investigated. AIMS In this study, 5-month-old APPswe/PS1dE9 transgenic mice were treated daily with 40 mg/kg artemisinin for 30 days by intraperitoneal injection to evaluate the effects of artemisinin on AD. RESULTS We found that artemisinin treatment (1) decreased neuritic plaque burden; (2) did not alter Aβ transport across the blood-brain barrier; (3) regulated APP processing via inhibiting β-secretase activity; (4) inhibited NF-κB activity and NALP3 inflammasome activation in APPswe/PS1dE9 double transgenic mice. CONCLUSIONS The in vivo study clearly demonstrates that artemisinin has protective effects on AD pathology due to its effects on suppressing NF-κB activity and NALP3 inflammasome activation. Our study suggests that targeting NF-κB activity and NALP3 inflammasome activation offers a valuable intervention for AD.
Collapse
Affiliation(s)
- Jian-Quan Shi
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Artemisinin suppresses sympathetic hyperinnervation following myocardial infarction via anti-inflammatory effects. J Mol Histol 2012; 43:737-43. [DOI: 10.1007/s10735-012-9440-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/11/2012] [Indexed: 11/26/2022]
|
20
|
Wen CC, Chen HM, Yang NS. Developing Phytocompounds from Medicinal Plants as Immunomodulators. ADVANCES IN BOTANICAL RESEARCH 2012; 62:197-272. [PMID: 32300254 PMCID: PMC7150268 DOI: 10.1016/b978-0-12-394591-4.00004-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Imbalance or malfunction of the immune systems is associated with a range of chronic diseases including autoimmune diseases, allergies, cancers and others. Various innate and adaptive immune cells that are integrated in this complex networking system may represent promising targets for developing immunotherapeutics for treating specific immune diseases. A spectrum of phytochemicals have been isolated, characterized and modified for development and use as prevention or treatment of human diseases. Many cytotoxic drugs and antibiotics have been developed from phytocompounds, but the application of traditional or new medicinal plants for use as immunomodulators in treating immune diseases is still relatively limited. In this review, a selected group of medicinal herbs, their derived crude or fractionated phytoextracts and the specific phytochemicals/phytocompounds isolated from them, as well as categorized phytocompound groups with specific chemical structures are discussed in terms of their immunomodulatory bioactivities. We also assess their potential for future development as immunomodulatory or inflammation-regulatory therapeutics or agents. New experimental approaches for evaluating the immunomodulatory activities of candidate phytomedicines are also discussed.
Collapse
Affiliation(s)
- Chih-Chun Wen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Hui-Ming Chen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Ning-Sun Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
21
|
Anti-inflammatory effect and modulation of cytochrome P450 activities by Artemisia annua tea infusions in human intestinal Caco-2 cells. Food Chem 2012; 134:864-71. [PMID: 23107701 DOI: 10.1016/j.foodchem.2012.02.195] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 02/16/2012] [Accepted: 02/28/2012] [Indexed: 12/11/2022]
Abstract
In an attempt to understand the beneficial health effects of Artemisia annua other than its anti-malaria properties, extracts from different cultivars prepared as tea infusions were investigated using Caco-2 cells on the intestinal inflammation and cytochrome P450 (CYP) activities. The characterisation of their phenolic compound (PC) profile revealed rosmarinic and chlorogenic acids as the main PCs. The extracts, assayed on Caco-2 cells at a plausible intestinal concentration, significantly decreased the secretion of pro-inflammatory cytokines, IL-8 and IL-6. This effect could be attributable at least to their content in rosmarinic acid, detected as a potent anti-inflammatory compound. The extracts also inhibited the activity of CYP3A4, whose expression was induced by 1,25-dihydroxyvitamin D(3), and of CYP1A1, induced by benzo(a)pyrene. Our results highlight the advantage of drinking A. annua infusions for their potent anti-inflammatory effect, linked to PC content, which could synergise their antimalarial activity.
Collapse
|