1
|
Liu X, Wang H, Lin D, Lin Z, Chen Q, Zhu B, Cao H. Multi-Auto-Tandem Reaction to Access Site-Specific Functionalized Tricyclic Furo[3,2- c]coumarins and Naphtho[2,3- b]furan-4,9-diones. Org Lett 2025. [PMID: 40271868 DOI: 10.1021/acs.orglett.5c00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
A three-component multi-auto-tandem reaction for the construction of site-specific tricyclic furo[3,2-c]coumarins via the formation of C-C, C-O, and C-S bonds in one step from 4-hydroxycoumarins/4-hydroxy-2-pyrones, ynals, and sodium sulfinates is reported. This cascade reaction efficiently produces a variety of rare C-2-functionalized furo[3,2-c]coumarins in moderate to good yields under straightforward reaction conditions. Furthermore, this protocol can be extended to a three-component coupling involving 2-hydroxy-1,4-naphthoquinone, ynals, and sodium sulfinates, yielding tricyclic naphtho[2,3-b]furan-4,9-dione derivatives. Notably, the carbonyl group and the α-position of ynals act as C-2 synthons in the specific multi-auto-tandem reaction, enabling the two aforementioned types of multicomponent transformations.
Collapse
Affiliation(s)
- Xiang Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| | - Hexiang Wang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| | - Dongrong Lin
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| | - Zhen Lin
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| | - Qiye Chen
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| | - Baofu Zhu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, P. R. China
| |
Collapse
|
2
|
Joshi A, Kathuria D, Paul M, Singh N. An overview on the potential application of nanotechnology in enhancing the therapeutic efficacy of phytoestrogens. Food Chem 2025; 464:141779. [PMID: 39481307 DOI: 10.1016/j.foodchem.2024.141779] [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: 05/23/2024] [Revised: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Phytoestrogens, derived from plants possesses structural similarity with 17 β-estradiol found in mammals. It is abundantly present in soybean along with red clove, alfalfa as well as other legumes, nuts, vegetables and seeds. It is used as hormone replacement therapy and exhibits both anti-estrogenic and estrogenic properties that linked to therapeutic benefits as well as plays active role in sports nutrition. Despite the potential benefits of phytoestrogens, their low solubility, bioavailability, and stability make it challenging to target them effectively. Recent advancements in nanotechnology have paved in facilitating target delivery. Scaling at nano level offered greater surface area, improved solubility, and bioavailability of phytoestrogens which has ultimately reduced the required medication dosage, and enhanced cost-effectiveness, particularly for expensive bioactive substances where precise dosages are recommended. The present article discussed about the potential application of nanotechnology in enhancing therapeutic benefits of phytoestrogens while minimizing their potential side effects.
Collapse
Affiliation(s)
- Aroma Joshi
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India
| | - Deepika Kathuria
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India
| | - Maman Paul
- Department of Physiotherapy, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Narpinder Singh
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India.
| |
Collapse
|
3
|
Faraz S, Ali A, Taleb Khan A. FeCl 3-catalyzed regioselective ring-opening of aryl oxirane with 4-hydroxycoumarin for the synthesis of furo[3,2- c]coumarins. Org Biomol Chem 2023; 22:95-105. [PMID: 38050428 DOI: 10.1039/d3ob01721d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
The regioselective ring-opening of aryl oxiranes was investigated with various 4-hydroxycoumarins in dimethyl sulfoxide in the presence of 20 mol% FeCl3 as a catalyst at 110 °C. This approach provided a short and concise synthetic route for the regioselective synthesis of 2-aryl-4H-furo[3,2-c] coumarin derivatives. Product formation occurred through regioselective ring-opening of the aryl oxirane at a less hindered site, followed by dehydration and concomitant cyclization. The salient features of our protocol were: cost-effectiveness; short reaction time; step- and atom economy; easy handling; broad scope of substrates; regioselectivity; good-to-excellent yields; non-requirement of dry solvents, co-catalysts, ligands, or any other additives; inert atmospheric conditions.
Collapse
Affiliation(s)
- Simra Faraz
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati-781039, India.
| | - Ahmad Ali
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati-781039, India.
| | - Abu Taleb Khan
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati-781039, India.
| |
Collapse
|
4
|
Liu C, Guo X, Zhou Y, Wang H. AMPK Signalling Pathway: A Potential Strategy for the Treatment of Heart Failure with Chinese Medicine. J Inflamm Res 2023; 16:5451-5464. [PMID: 38026240 PMCID: PMC10676094 DOI: 10.2147/jir.s441597] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome that represents the advanced stage of cardiovascular disease, characterized by systolic and diastolic dysfunction of the heart. Despite continuous updates in HF treatment drugs, the morbidity and mortality rates remain high, necessitating ongoing exploration for new therapeutic targets. Adenosine monophosphate-activated protein kinase (AMPK) is the serine/threonine protein kinase which responds to adenosine monophosphate (AMP) levels.Activation of AMPK shifts cellular metabolic patterns from synthesis to catabolism, enhancing energy metabolism in pathological conditions such as inflammation, ischemia, obesity, and aging. Numerous studies have identified AMPK as a vital target for HF treatment, with herbal monomers/extracts and compounds affecting key signaling factors including rapamycin targeting protein (mTOR), silencing regulator protein 1 (SIRT1), nuclear transcription factor E2-related factor 2 (Nrf2), and nuclear transcription factor-κB (NF-κB) through regulation of the AMPK signaling pathway.This modulation can achieve the effects of improving metabolism, autophagy, reducing oxidative stress and inflammatory response in the treatment of heart failure, with the advantages of multi-targeting, comprehensive action and low toxicity.The modulation of the AMPK pathway by Traditional Chinese Medicine (TCM) has emerged as a crucial research direction for the prevention and treatment of HF, but a systematic summary and generalization in this field is lacking. This article provides an overview of the composition, regulation, and mechanism of the AMPK signaling pathway's influence on HF, as well as a summary of current research on the regulation of the AMPK pathway by TCM for HF prevention and treatment. The aim is to serve as a reference for the diagnosis and treatment of HF using TCM and the development of new drugs.
Collapse
Affiliation(s)
- Changxing Liu
- First Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| | - Xinyi Guo
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People’s Republic of China
| | - Yabin Zhou
- Department of Cardiology, The First Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| | - He Wang
- Department of Cardiology, The First Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| |
Collapse
|
5
|
Yang L, Guan J, Luo S, Yan J, Chen D, Zhang X, Zhong C, Yang P. Angiotensin IV ameliorates doxorubicin-induced cardiotoxicity by increasing glutathione peroxidase 4 and alleviating ferroptosis. Toxicol Appl Pharmacol 2023; 479:116713. [PMID: 37838222 DOI: 10.1016/j.taap.2023.116713] [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: 07/04/2023] [Revised: 09/20/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Doxorubicin (DOX)-induced cardiotoxicity is an important cause of poor prognosis in cancer patients treated with DOX. Angiotensin IV (Ang IV) has multiple protective effects against cardiovascular diseases, including diabetic cardiomyopathy and myocardial infarction, but its role in DOX-induced cardiotoxicity is currently unclear. In this study, we investigated the effects of Ang IV on DOX-induced cardiotoxicity. METHODS The viability of primary cardiomyocytes was measured by Cell Counting Kit-8 assays and Hoechst 33342/propidium iodide staining in vitro. ELISAs (serum cTnT and CK-MB) and echocardiography were performed to assess myocardial injury and cardiac function in vivo. Phalloidin staining, haematoxylin and eosin staining and wheat germ agglutinin staining were conducted to detect cardiomyocyte atrophy. We also performed C11 BODIPY staining, measured the levels of Ptgs2 and malondialdehyde and detected the concentrations of ferrous ions, glutathione and oxidized glutathione to indicate ferroptosis. RESULTS Ang IV not only attenuated DOX-induced atrophy and cardiomyocyte injury in vitro but also alleviated myocardial injury and improved cardiac function in DOX-treated mice in vivo. Moreover, Ang IV reversed DOX-induced downregulation of glutathione peroxidase 4 (GPX4) and inhibited ferroptosis both in vitro and in vivo. Knockdown of GPX4 by siRNA abolished the cardioprotective effects of Ang IV. Furthermore, Ang IV increased GPX4 levels and ameliorated ferroptosis in RAS-selective lethal 3-treated primary cardiomyocytes. CONCLUSIONS Ang IV ameliorates DOX-induced cardiotoxicity by upregulating GPX4 and inhibiting ferroptosis. Ang IV may be a promising candidate to protect against DOX-induced cardiotoxicity in the future.
Collapse
Affiliation(s)
- Li Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China; Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, Hunan, People's Republic of China
| | - Junjie Guan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China
| | - Shen Luo
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, Hunan, People's Republic of China
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China
| | - Deshu Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China
| | - Xuwei Zhang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China
| | - Chongbin Zhong
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China.
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, People's Republic of China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Sun Y, Guo D, Yue S, Zhou M, Wang D, Chen F, Wang L. Afzelin protects against doxorubicin-induced cardiotoxicity by promoting the AMPKα/SIRT1 signaling pathway. Toxicol Appl Pharmacol 2023; 477:116687. [PMID: 37703929 DOI: 10.1016/j.taap.2023.116687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Doxorubicin (DOX), a chemotherapeutic drug, could relieve the progressions of various diseases. However, its clinical application is limited due to its cardiotoxicity. This study aimed to investigate the effects of afzelin (a flavonol glycoside found in Houttuynia cordata) on the cardiotoxicity induced by DOX. METHODS In ex-vivo, H9C2 cells were incubated with 20, 40, or 80 μM afzelin for 12 h, followed by the treatment with 1 μM DOX for 12 h. In vivo, C57BL/6 J mice were intraperitoneally injected with 4 mg/kg/day DOX on days 1, 7, and 14. Meanwhile, starting from day 1, mice were intragastrically administrated with 5 mg/kg/day or 10 mg/kg/day afzelin for 20 days. The cardiac function of mice was evaluated by detecting hemodynamic parameters using the M-mode echocardiography. RESULTS DOX decreased the cell survival rate, and elevated apoptotic rate, as well as induced the oxidative stress and mitochondrial dysfunction in H9C2 cells. All these changes were alleviated by afzelin treatment in a concentration-dependent manner. The results were further proven by the mitigation of cardiac injury in vivo, as evidenced by the elevation of fractional shortening, heart weight/tibia length, and the rate of the increase/decrease of left ventricular pressure in mice subjected to DOX-induced cardiotoxicity. Furthermore, afzelin upregulated the expression of p-AMP-activated protein kinase alpha (AMPKα) and sirtuin1 (SIRT1). Dorsomorphin (an AMPKα inhibitor) abrogated the anti-cardiotoxicity effects of afzelin in H9C2 cells induced by DOX. CONCLUSION Afzelin protected against DOX-induced cardiotoxicity by promoting the AMPKα/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Yixin Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Danyang Guo
- Department of Ultrasound, the Sixth Affiliated Hospital of Harbin Medical University, 57 Youyi Road, Daoli District, Harbin 150076, Heilongjiang, China
| | - Saiding Yue
- Department of Nephrology, Harbin Jing-En Nephrology Hospital, 11 Xiangbin Road, Xiangfang District, Harbin 150036, Heilongjiang, China
| | - Mingyan Zhou
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Dongxu Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Fengjiao Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Lingling Wang
- Department of Ultrasound, the Sixth Affiliated Hospital of Harbin Medical University, 998 Aiying Avenue, Songbei District, Harbin 150027, Heilongjiang, China.
| |
Collapse
|
7
|
Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother 2022; 156:113903. [PMID: 36279722 DOI: 10.1016/j.biopha.2022.113903] [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: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/06/2022] Open
Abstract
Doxorubicin (DOX), as a kind of chemotherapy agent with remarkable therapeutic effect, can be used to treat diverse malignant tumors clinically. Dose-dependent cardiotoxicity is the most serious adverse reaction after DOX treatment, which eventually leads to cardiomyopathy and greatly limits the clinical application of DOX. DOX-induced cardiomyopathy is not a result of a single mechanistic action, and multiple mechanisms have been discovered and demonstrated experimentally, such as oxidative stress, inflammation, mitochondrial damage, calcium homeostasis disorder, ferroptosis, autophagy and apoptosis. Dexrazoxane (DEX) is the only protective agent approved by FDA for the treatment of DOX cardiomyopathy, but its clinical treatment still has some limitations. Therefore, we need to find other effective therapeutic drugs as soon as possible. In this paper, the drugs that effectively improve cardiomyopathy in recent years are mainly described from the aspects of natural drugs, endogenous substances, new dosage forms, herbal medicines, chemical modification and marketed drugs. The aim of the present study is to evaluate the effects of these drugs on DOX-induced anticancer and cardiomyopathy curative effects, so as to provide some reference value for clinical treatment of DOX-induced cardiomyopathy in the future.
Collapse
Affiliation(s)
- Ye Chen
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China; School of pharmacy, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Saixian Shi
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China; School of pharmacy, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yan Dai
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
8
|
Wu X, Shen F, Jiang G, Xue G, Philips S, Gardner L, Cunningham G, Bales C, Cantor E, Schneider BP. A non-coding GWAS variant impacts anthracycline-induced cardiotoxic phenotypes in human iPSC-derived cardiomyocytes. Nat Commun 2022; 13:7171. [PMID: 36418322 PMCID: PMC9684507 DOI: 10.1038/s41467-022-34917-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/11/2022] [Indexed: 11/24/2022] Open
Abstract
Anthracyclines, widely used to treat breast cancer, have the potential for cardiotoxicity. We have previously identified and validated a germline single nucleotide polymorphism, rs28714259, associated with an increased risk of anthracycline-induced heart failure. We now provide insights into the mechanism by which rs28714259 might confer increased risk of cardiac damage. Using hiPSC-derived cardiomyocyte cell lines with either intrinsic polymorphism or CRISPR-Cas9-mediated deletion of rs28714259 locus, we demonstrate that glucocorticoid receptor signaling activated by dexamethasone pretreatment prior to doxorubicin exposure preserves cardiomyocyte viability and contractility in cardiomyocytes containing the major allele. Homozygous loss of the rs28714259 major allele diminishes dexamethasone's protective effect. We further demonstrate that the risk allele of rs28714259 disrupts glucocorticoid receptor and rs28714259 binding affinity. Finally, we highlight the activation of genes and pathways involved in cardiac hypertrophy signaling that are blocked by the risk allele, suggesting a decreased adaptive survival response to doxorubicin-related stress.
Collapse
Affiliation(s)
- Xi Wu
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fei Shen
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Guanglong Jiang
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gloria Xue
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Santosh Philips
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Laura Gardner
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Geneva Cunningham
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Casey Bales
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Erica Cantor
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bryan Paul Schneider
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
9
|
Aiyasiding X, Liao HH, Feng H, Zhang N, Lin Z, Ding W, Yan H, Zhou ZY, Tang QZ. Liquiritin Attenuates Pathological Cardiac Hypertrophy by Activating the PKA/LKB1/AMPK Pathway. Front Pharmacol 2022; 13:870699. [PMID: 35592411 PMCID: PMC9110825 DOI: 10.3389/fphar.2022.870699] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Liquiritin (LQ) is one of the main flavonoids extracted from the roots of Glycyrrhiza spp., which are widely used in traditional Chinese medicine. Studies in both cellular and animal disease models have shown that LQ attenuates or prevents oxidative stress, inflammation, and apoptosis. However, the potential therapeutic effects of LQ on pressure overload-induced cardiac hypertrophy have not been so far explored. Therefore, we investigated the cardioprotective role of LQ and its underlying mechanisms in the aortic banding (AB)-induced cardiac hypertrophy mouse model. Methods and Results: Starting 3 days after AB surgery, LQ (80 mg/kg/day) was administered daily over 4 weeks. Echocardiography and pressure-volume loop analysis indicated that LQ treatment markedly improved hypertrophy-related cardiac dysfunction. Moreover, hematoxylin and eosin, picrosirius red, and TUNEL staining showed that LQ significantly inhibited cardiomyocyte hypertrophy, interstitial fibrosis, and apoptosis. Western blot assays further showed that LQ activated LKB1/AMPKα2/ACC signaling and inhibited mTORC1 phosphorylation in cardiomyocytes. Notably, LQ treatment failed to prevent cardiac dysfunction, hypertrophy, and fibrosis in AMPKα2 knockout (AMPKα2−/−) mice. However, LQ still induced LKB1 phosphorylation in AMPKα2−/− mouse hearts. In vitro experiments further demonstrated that LQ inhibited Ang II-induced hypertrophy in neonatal rat cardiomyocytes (NRCMs) by increasing cAMP levels and PKA activity. Supporting the central involvement of the cAMP/PKA/LKB1/AMPKα2 signaling pathway in the cardioprotective effects of LQ, inhibition of Ang II-induced hypertrophy and induction of LKB1 and AMPKα phosphorylation were no longer observed after inhibiting PKA activity. Conclusion: This study revealed that LQ alleviates pressure overload-induced cardiac hypertrophy in vivo and inhibits Ang II-induced cardiomyocyte hypertrophy in vitro via activating cAMP/PKA/LKB1/AMPKα2 signaling. These findings suggest that LQ might be a valuable adjunct to therapeutic approaches for treating pathological cardiac remodeling.
Collapse
Affiliation(s)
- Xiahenazi Aiyasiding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Hai-Han Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Hong Feng
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Zheng Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Han Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Zi-Ying Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| |
Collapse
|
10
|
Adhikari A, Asdaq SMB, Al Hawaj MA, Chakraborty M, Thapa G, Bhuyan NR, Imran M, Alshammari MK, Alshehri MM, Harshan AA, Alanazi A, Alhazmi BD, Sreeharsha N. Anticancer Drug-Induced Cardiotoxicity: Insights and Pharmacogenetics. Pharmaceuticals (Basel) 2021; 14:ph14100970. [PMID: 34681194 PMCID: PMC8539940 DOI: 10.3390/ph14100970] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
The advancement in therapy has provided a dramatic improvement in the rate of recovery among cancer patients. However, this improved survival is also associated with enhanced risks for cardiovascular manifestations, including hypertension, arrhythmias, and heart failure. The cardiotoxicity induced by chemotherapy is a life-threatening consequence that restricts the use of several chemotherapy drugs in clinical practice. This article addresses the prevalence of cardiotoxicity mediated by commonly used chemotherapeutic and immunotherapeutic agents. The role of susceptible genes and radiation therapy in the occurrence of cardiotoxicity is also reviewed. This review also emphasizes the protective role of antioxidants and future perspectives in anticancer drug-induced cardiotoxicities.
Collapse
Affiliation(s)
- Archana Adhikari
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Syed Mohammed Basheeruddin Asdaq
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, Riyadh 13713, Saudi Arabia
- Correspondence: (S.M.B.A.); (M.C.)
| | - Maitham A. Al Hawaj
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Hofuf 31982, Saudi Arabia;
| | - Manodeep Chakraborty
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
- Correspondence: (S.M.B.A.); (M.C.)
| | - Gayatri Thapa
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Nihar Ranjan Bhuyan
- Department of Pharmaceutical Analysis, Himalayan Pharmacy Institute, Majhitar, Rangpo 737136, Sikkim, India;
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | | | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Aishah Ali Harshan
- Department of Pharmaceutical Care, Northern Area Armed Forces Hospital, King Khalid Military City Hospital, Hafr Al-Batin 39745, Saudi Arabia;
| | - Abeer Alanazi
- Department of Pharmaceutical Care, First Health Cluster in Eastern Province, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia;
| | | | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa-31982, Saudi Arabia;
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bengaluru 560035, Karnataka, India
| |
Collapse
|
11
|
Chen Z, Zeng P, Zhang S, Huang X. Lewis‐Acid‐Mediated One‐Pot Tandem Reactions for Synthesis of Structurally Diverse Furo[3,2‐c]coumarins. ChemistrySelect 2021. [DOI: 10.1002/slct.202101029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zhiwei Chen
- College of Pharmaceutical Sciences Zhejiang University of Technology Hangzhou 310014 P.R. China
| | - Piaopiao Zeng
- College of Pharmaceutical Sciences Zhejiang University of Technology Hangzhou 310014 P.R. China
| | - Shuo Zhang
- College of Pharmaceutical Sciences Zhejiang University of Technology Hangzhou 310014 P.R. China
| | - Xiaoxiao Huang
- College of Pharmaceutical Sciences Zhejiang University of Technology Hangzhou 310014 P.R. China
| |
Collapse
|