1
|
He M, Wu H, Xu T, Zhao Y, Wang Z, Liu Y. Fangchinoline eliminates intracellular Salmonella by enhancing lysosomal function via the AMPK-mTORC1-TFEB axis. J Adv Res 2025:S2090-1232(25)00034-7. [PMID: 39788287 DOI: 10.1016/j.jare.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Salmonella, a foodborne zoonotic pathogen, is a significant cause of morbidity and mortality in animals and humans globally. With the prevalence of multidrug-resistant strains, Salmonellosis has become a formidable challenge. Host-directed therapy (HDT) has recently emerged as a promising anti-infective approach for treating intracellular bacterial infections. OBJECTIVES Plant-derived natural products, owing to their structural and functional diversity, are increasingly being explored and utilized as encouraging candidates for HDT compounds. This study aims to identify and screen natural compounds with potential as HDT for the treatment of intracellular Salmonella infections. METHODS A cell-based screening approach was deployed to identify natural compounds capable of mitigating the intracellular replication of S. enterica. Safety and efficacy of the candidate compounds were evaluated using multiple animal models. RNA sequencing, ELISA, and immunoblotting analyses were conducted to elucidate the underlying mechanisms of action. RESULTS Our results reveal that fangchinoline (FAN) effectively reduces S. enterica survival both in vitro and in vivo. Meanwhile, FAN also displays anti-infective activity against other intracellular pathogens, including multidrug-resistant isolates. A 14-day safety evaluation in mice showed no significant toxic or adverse effects from FAN administration. RNA sequencing analysis reveals an upregulation of lysosome pathways in S. enterica-infected cells treated with FAN. Mechanistic studies indicate that FAN increases acid lysosomal quantities and fosters autophagic response in Salmonella-infected cells via the AMPK-mTORC1-TFEB axis. In addition, FAN alleviates the inflammatory response in Salmonella-infected cells by inactivating the NF-κB pathway. CONCLUSION Our findings suggest that FAN represents a lead HDT compound for tackling recalcitrant infections caused by intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Mengping He
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Huihui Wu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yurong Zhao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China.
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.
| |
Collapse
|
2
|
Huang L, Liu L, Zhu J, Chen N, Chen J, Chan CF, Gao F, Yin Y, Sun J, Zhang R, Zhang K, Qi W, Yue J. Bis-benzylisoquinoline alkaloids inhibit flavivirus entry and replication by compromising endolysosomal trafficking and autophagy. Virol Sin 2024; 39:892-908. [PMID: 39251138 PMCID: PMC11738800 DOI: 10.1016/j.virs.2024.09.001] [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: 05/26/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Flaviviruses, such as dengue virus (DENV), Zika virus (ZIKV), and Japanese encephalitis virus (JEV), represent a substantial public health challenge as there are currently no approved treatments available. Here, we investigated the antiviral effects of bis-benzylisoquinoline alkaloids (BBAs) on flavivirus infections. We evaluated five specific BBAs-berbamine, tetrandrine, iso-tetrandrine, fangchinoline, and cepharanthine-and found that they effectively inhibited infections by ZIKV, DENV, or JEV by blocking virus entry and genome replication stages in the flavivirus life cycle. Furthermore, we synthesized a fluorophore-conjugated BBA and showed that BBAs targeted endolysosomes, causing lysosomal pH alkalization. Mechanistic studies on inhibiting ZIKV infection by BBAs revealed that these compounds blocked TRPML channels, leading to lysosomal dysfunction and reducing the expression of NCAM1, a key receptor for the entry of ZIKV into cells, thereby decreasing cells susceptibility to ZIKV infection. Additionally, BBAs inhibited the fusion of autophagosomes and lysosomes, significantly reducing viral RNA replication. Collectively, our results suggest that BBAs inhibit flavivirus entry and replication by compromising endolysosomal trafficking and autophagy, respectively, underscoring the potential of BBAs as therapeutic agents against flavivirus infections.
Collapse
Affiliation(s)
- Lihong Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Lele Liu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Junhai Zhu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Nanjun Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong, 999077, China
| | - Jie Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China
| | - Chuen-Fuk Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, 999077, China
| | - Fei Gao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Youqin Yin
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Jiufeng Sun
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Kehui Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Wenbao Qi
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China.
| | - Jianbo Yue
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China; Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Global Health Research Center, Duke Kunshan University, Kunshan, 215316, China; College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
3
|
Su G, Yang X, Lin Q, Su G, Liu J, Huang L, Chen W, Wei W, Chen J. Fangchinoline Inhibits African Swine Fever Virus Replication by Suppressing the AKT/mTOR/NF-κB Signaling Pathway in Porcine Alveolar Macrophages. Int J Mol Sci 2024; 25:7178. [PMID: 39000284 PMCID: PMC11241579 DOI: 10.3390/ijms25137178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 07/16/2024] Open
Abstract
African swine fever (ASF), caused by the African swine fever virus (ASFV), is one of the most important infectious diseases that cause high morbidity and mortality in pigs and substantial economic losses to the pork industry of affected countries due to the lack of effective vaccines. The need to develop alternative robust antiviral countermeasures, especially anti-ASFV agents, is of the utmost urgency. This study shows that fangchinoline (FAN), a bisbenzylisoquinoline alkaloid found in the roots of Stephania tetrandra of the family Menispermaceae, significantly inhibits ASFV replication in porcine alveolar macrophages (PAMs) at micromolar concentrations (IC50 = 1.66 µM). Mechanistically, the infection of ASFV triggers the AKT/mTOR/NF-κB signaling pathway. FAN significantly inhibits ASFV-induced activation of such pathways, thereby suppressing viral replication. Such a mechanism was confirmed using an AKT inhibitor MK2206 as it inhibited AKT phosphorylation and ASFV replication in PAMs. Altogether, the results suggest that the AKT/mTOR pathway could potentially serve as a treatment strategy for combating ASFV infection and that FAN could potentially emerge as an effective novel antiviral agent against ASFV infections and deserves further in vivo antiviral evaluations.
Collapse
Affiliation(s)
- Guanming Su
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoqun Yang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Qisheng Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Guoming Su
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jinyi Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Li Huang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Wenkang Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, Agro-Biological Gene Research Center of Guangdong Academy of Agricultural Sciences, Guangzhou 510642, China
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
4
|
Wang J, Xie F, Jia X, Wang X, Kong L, Li Y, Liang X, Zhang M, He Y, Feng W, Luo T, Wang Y, Xu A. Fangchinoline induces antiviral response by suppressing STING degradation. J Pharm Anal 2024; 14:100972. [PMID: 39027910 PMCID: PMC11255895 DOI: 10.1016/j.jpha.2024.100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 07/20/2024] Open
Abstract
The stimulator of interferon genes (STING), an integral adaptor protein in the DNA-sensing pathway, plays a pivotal role in the innate immune response against infections. Additionally, it presents a valuable therapeutic target for infectious diseases and cancer. We observed that fangchinoline (Fan), a bis-benzylisoquinoline alkaloid (BBA), effectively impedes the replication of vesicular stomatitis virus (VSV), encephalomyocarditis virus (EMCV), influenza A virus (H1N1), and herpes simplex virus-1 (HSV-1) in vitro. Fan treatment significantly reduced the viral load, attenuated tissue inflammation, and improved survival in a viral sepsis mouse model. Mechanistically, Fan activates the antiviral response in a STING-dependent manner, leading to increased expression of interferon (IFN) and interferon-stimulated genes (ISGs) for potent antiviral effects in vivo and in vitro. Notably, Fan interacts with STING, preventing its degradation and thereby extending the activation of IFN-based antiviral responses. Collectively, our findings highlight the potential of Fan, which elicits antiviral immunity by suppressing STING degradation, as a promising candidate for antiviral therapy.
Collapse
Affiliation(s)
- Jinyong Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fang Xie
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuejiao Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lingdong Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yiying Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue Liang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Meiqi Zhang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yuting He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wandi Feng
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Tong Luo
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yao Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Anlong Xu
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
5
|
Yang L, Wang X, Ma Z, Sui Y, Liu X. Fangchinoline inhibits growth and biofilm of Candida albicans by inducing ROS overproduction. J Cell Mol Med 2024; 28:e18354. [PMID: 38686557 PMCID: PMC11058694 DOI: 10.1111/jcmm.18354] [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: 12/31/2023] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Infections caused by Candida species, especially Candida albicans, threaten the public health and create economic burden. Shortage of antifungals and emergence of drug resistance call for new antifungal therapies while natural products were attractive sources for developing new drugs. In our study, fangchinoline, a bis-benzylisoquinoline alkaloid from Chinese herb Stephania tetrandra S. Moore, exerted antifungal effects on planktonic growth of several Candida species including C. albicans, with MIC no more than 50 μg/mL. In addition, results from microscopic, MTT and XTT reduction assays showed that fangchinoline had inhibitory activities against the multiple virulence factors of C. albicans, such as adhesion, hyphal growth and biofilm formation. Furthermore, this compound could also suppress the metabolic activity of preformed C. albicans biofilms. PI staining, followed by confocal laser scanning microscope (CLSM) analysis showed that fangchinoline can elevate permeability of cell membrane. DCFH-DA staining suggested its anti-Candida mechanism also involved overproduction of intracellular ROS, which was further confirmed by N-acetyl-cysteine rescue tests. Moreover, fangchinoline showed synergy with three antifungal drugs (amphotericin B, fluconazole and caspofungin), further indicating its potential use in treating C. albicans infections. Therefore, these results indicated that fangchinoline could be a potential candidate for developing anti-Candida therapies.
Collapse
Affiliation(s)
- Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical GeneticsThe Second Hospital of Jilin UniversityChangchunChina
| | - Xiaonan Wang
- Department of OrthopedicsThe Second Hospital of Jilin UniversityChangchunChina
| | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia SurgeryThe Second Hospital of Jilin UniversityChangchunChina
| | - Yujie Sui
- Jilin Provincial Key Laboratory on Molecular and Chemical GeneticsThe Second Hospital of Jilin UniversityChangchunChina
| | - Xin Liu
- Eye Center, The Second Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
6
|
Zhang QY, Li JQ, Li Q, Zhang Y, Zhang ZR, Li XD, Zhang HQ, Deng CL, Yang FX, Xu Y, Zhang B. Identification of fangchinoline as a broad-spectrum enterovirus inhibitor through reporter virus based high-content screening. Virol Sin 2024; 39:301-308. [PMID: 38452856 DOI: 10.1016/j.virs.2024.02.006] [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: 07/05/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Hand, foot, and mouth disease (HFMD) is a common pediatric illness mainly caused by enteroviruses, which are important human pathogens. Currently, there are no available antiviral agents for the therapy of enterovirus infection. In this study, an excellent high-content antiviral screening system utilizing the EV-A71-eGFP reporter virus was developed. Using this screening system, we screened a drug library containing 1042 natural compounds to identify potential EV-A71 inhibitors. Fangchinoline (FAN), a bis-benzylisoquinoline alkaloid, exhibits potential inhibitory effects against various enteroviruses that cause HFMD, such as EV-A71, CV-A10, CV-B3 and CV-A16. Further investigations revealed that FAN targets the early stage of the enterovirus life cycle. Through the selection of FAN-resistant EV-A71 viruses, we demonstrated that the VP1 protein could be a potential target of FAN, as two mutations in VP1 (E145G and V258I) resulted in viral resistance to FAN. Our research suggests that FAN is an efficient inhibitor of EV-A71 and has the potential to be a broad-spectrum antiviral drug against human enteroviruses.
Collapse
Affiliation(s)
- Qiu-Yan Zhang
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jia-Qi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yang Zhang
- University of Science and Technology of China, Department of Life Sciences and Medicine, Hefei, 230026, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiao-Dan Li
- Hunan Normal University, School of Medicine, Changsha, 410081, China
| | - Hong-Qing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Feng-Xia Yang
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
| | - Yi Xu
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China.
| | - Bo Zhang
- The Joint Center of Translational Precision Medicine, Department of Infections and Diseases, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
7
|
Shah M, Yamin R, Ahmad I, Wu G, Jahangir Z, Shamim A, Nawaz H, Nishan U, Ullah R, Ali EA, Sheheryar, Chen K. In-silico evaluation of natural alkaloids against the main protease and spike glycoprotein as potential therapeutic agents for SARS-CoV-2. PLoS One 2024; 19:e0294769. [PMID: 38175855 PMCID: PMC10766191 DOI: 10.1371/journal.pone.0294769] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/08/2023] [Indexed: 01/06/2024] Open
Abstract
Severe Acute Respiratory Syndrome Corona Virus (SARS-CoV-2) is the causative agent of COVID-19 pandemic, which has resulted in global fatalities since late December 2019. Alkaloids play a significant role in drug design for various antiviral diseases, which makes them viable candidates for treating COVID-19. To identify potential antiviral agents, 102 known alkaloids were subjected to docking studies against the two key targets of SARS-CoV-2, namely the spike glycoprotein and main protease. The spike glycoprotein is vital for mediating viral entry into host cells, and main protease plays a crucial role in viral replication; therefore, they serve as compelling targets for therapeutic intervention in combating the disease. From the selection of alkaloids, the top 6 dual inhibitory compounds, namely liensinine, neferine, isoliensinine, fangchinoline, emetine, and acrimarine F, emerged as lead compounds with favorable docked scores. Interestingly, most of them shared the bisbenzylisoquinoline alkaloid framework and belong to Nelumbo nucifera, commonly known as the lotus plant. Docking analysis was conducted by considering the key active site residues of the selected proteins. The stability of the top three ligands with the receptor proteins was further validated through dynamic simulation analysis. The leads underwent ADMET profiling, bioactivity score analysis, and evaluation of drug-likeness and physicochemical properties. Neferine demonstrated a particularly strong affinity for binding, with a docking score of -7.5025 kcal/mol for main protease and -10.0245 kcal/mol for spike glycoprotein, and therefore a strong interaction with both target proteins. Of the lead alkaloids, emetine and fangchinoline demonstrated the lowest toxicity and high LD50 values. These top alkaloids, may support the body's defense and reduce the symptoms by their numerous biological potentials, even though some properties naturally point to their direct antiviral nature. These findings demonstrate the promising anti-COVID-19 properties of the six selected alkaloids, making them potential candidates for drug design. This study will be beneficial in effective drug discovery and design against COVID-19 with negligible side effects.
Collapse
Affiliation(s)
- Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Ramsha Yamin
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Iqra Ahmad
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Gang Wu
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zainab Jahangir
- Department of Computer Science, University of Agriculture Faisalabad, Punjab, Pakistan
| | - Amen Shamim
- Department of Computer Science, University of Agriculture Faisalabad, Punjab, Pakistan
| | - Haq Nawaz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Umar Nishan
- Department of Chemistry, Kohat University of Science & Technology, Kohat, Pakistan
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Essam A. Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheheryar
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, Fortaleza, Brazil
| | - Ke Chen
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
8
|
Sadhu S, Dandotiya J, Dalal R, Khatri R, Mykytyn AZ, Batra A, Kaur M, Chandwaskar R, Singh V, Kamboj A, Srivastava M, Mani S, Asthana S, Samal S, Rizvi ZA, Salunke DB, Haagmans BL, Awasthi A. Fangchinoline inhibits SARS-CoV-2 and MERS-CoV entry. Antiviral Res 2023; 220:105743. [PMID: 37949319 DOI: 10.1016/j.antiviral.2023.105743] [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: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2, lead to mild to severe respiratory illness and resulted in 6.9 million deaths worldwide. Although vaccines are effective in preventing COVID-19, they may not be sufficient to protect immunocompromised individuals from this respiratory illness. Moreover, novel emerging variants of SARS-CoV-2 pose a risk of new COVID-19 waves. Therefore, identification of effective antivirals is critical in controlling SARS and other coronaviruses, such as MERS-CoV. We show that Fangchinoline (Fcn), a bisbenzylisoquinoline alkaloid, inhibits replication of SARS-CoV, SARS-CoV-2, and MERS-CoV in a range of in vitro assays, by blocking entry. Therapeutic use of Fcn inhibited viral loads in the lungs, and suppressed associated airway inflammation in hACE2. Tg mice and Syrian hamster infected with SARS-CoV-2. Combination of Fcn with remdesivir (RDV) or an anti-leprosy drug, Clofazimine, exhibited synergistic antiviral activity. Compared to Fcn, its synthetic derivative, MK-04-003, more effectively inhibited SARS-CoV-2 and its variants B.1.617.2 and BA.5 in mice. Taken together these data demonstrate that Fcn is a pan beta coronavirus inhibitor, which possibly can be used to combat novel emerging coronavirus diseases.
Collapse
Affiliation(s)
- Srikanth Sadhu
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India; Immunology-Core Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Jyotsna Dandotiya
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Rajdeep Dalal
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Ritika Khatri
- Infection and Immunology Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Anna Z Mykytyn
- Viroscience Department, Erasmus University Medical Center, Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Netherlands
| | - Aashima Batra
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Manpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | | | - Virendra Singh
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Aarzoo Kamboj
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Mitul Srivastava
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Shailendra Mani
- Infection and Immunology Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Shailendra Asthana
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Sweety Samal
- Infection and Immunology Center, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Zaigham Abbas Rizvi
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India; Immunology-Core Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Bart L Haagmans
- Viroscience Department, Erasmus University Medical Center, Netherlands
| | - Amit Awasthi
- Center for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India; Immunology-Core Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
9
|
Zhang W, Shen H, Wang M, Fan X, Wang S, Wuri N, Zhang B, He H, Zhang C, Liu Z, Liao M, Zhang J, Li Y, Zhang J. Fangchinoline inhibits the PEDV replication in intestinal epithelial cells via autophagic flux suppression. Front Microbiol 2023; 14:1164851. [PMID: 37485535 PMCID: PMC10360400 DOI: 10.3389/fmicb.2023.1164851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Animal and human health are severely threatened by coronaviruses. The enteropathogenic coronavirus, porcine epidemic diarrhea virus (PEDV), is highly contagious, leading to porcine epidemic diarrhea (PED), which causes large economic losses in the world's swine industry. Piglets are not protected from emerging PEDV variants; therefore, new antiviral measures for PED control are urgently required. Herein, the anti-PEDV effects and potential mechanisms of fangchinoline (Fan) were investigated. Fan dose-dependently inhibited a PEDV infection at 24 h post-infection (EC50 value = 0.67 μM). We found that Fan mainly affected the PEDV replication phase but also inhibited PEDV at the attachment and internalization stages of the viral life cycle. Mechanistically, Fan blocked the autophagic flux in PEDV-infected cells by regulating the expression of autophagy-related proteins and changing PEDV virus particles. In summary, Fan inhibits PEDV infection by blocking the autophagic flux in cells. Our findings will help develop new strategies to prevent and treat PEDV infection.
Collapse
Affiliation(s)
- Weixiao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Haiyan Shen
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Guangzhou, China
| | - Menglu Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xuelei Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Songqi Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Nile Wuri
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Bin Zhang
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Haiyan He
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Chunhong Zhang
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Guangzhou, China
| | - Zhicheng Liu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Guangzhou, China
| | - Ming Liao
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Guangzhou, China
| | - Jianfeng Zhang
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Guangzhou, China
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jianmin Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
10
|
Zhang C, Chen H, Sun L, Zhao P, Qi C, Yang Y, Si A, Qian Y, Jung YS. Bis-Benzylisoquinoline Alkaloids Inhibit Porcine Epidemic Diarrhea Virus by Disrupting Virus Entry. Pathogens 2023; 12:845. [PMID: 37375535 DOI: 10.3390/pathogens12060845] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The porcine epidemic diarrhea virus (PEDV), belonging to the α-coronavirus, is the causative agent of porcine epidemic diarrhea (PED). Presently, protection from the existing PEDV vaccine is not effective. Therefore, anti-PEDV compounds should be studied. Berbamine (BBM), Fangchinoline (FAN), and (+)-Fangchinoline (+FAN), are types of bis-benzylisoquinoline alkaloids that are extracted from natural medicinal plants. These bis-benzylisoquinoline alkaloids have various biological activities, including antiviral, anticancer, and anti-inflammatory properties. In this study, we found that BBM, FAN, and +FAN suppressed PEDV activity with a 50% inhibitory concentration of 9.00 µM, 3.54 µM, and 4.68 µM, respectively. Furthermore, these alkaloids can decrease the PEDV-N protein levels and virus titers in vitro. The time-of-addition assay results showed that these alkaloids mainly inhibit PEDV entry. We also found that the inhibitory effects of BBM, FAN, and +FAN on PEDV rely on decreasing the activity of Cathepsin L (CTSL) and Cathepsin B (CTSB) by suppressing lysosome acidification. Taken together, these results indicated that BBM, FAN, and +FAN were effective anti-PEDV natural products that prevented PEDV entry and may be considered novel antiviral drugs.
Collapse
Affiliation(s)
- Caisheng Zhang
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Huan Chen
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Liumei Sun
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Pu Zhao
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chuanxiang Qi
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ying Yang
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Anqi Si
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yingjuan Qian
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Veterinary Bio-Pharmaceutical, Taizhou 225300, China
| | - Yong-Sam Jung
- One Health Laboratory, Jiangsu Foreign Expert Workshop, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
11
|
Faisal S, Badshah SL, Kubra B, Emwas AH, Jaremko M. Alkaloids as potential antivirals. A comprehensive review. NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:4. [PMID: 36598588 PMCID: PMC9812014 DOI: 10.1007/s13659-022-00366-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/01/2022] [Indexed: 05/26/2023]
Abstract
Alkaloids are a diverse group of natural phytochemicals. These phytochemicals in plants provide them protection against pests, and herbivorous organisms and also control their development. Numerous of these alkaloids have a variety of biological effects, and some have even been developed into medications with different medicinal properties. This review aims to provide a broad overview of the numerous naturally occurring alkaloids (isolated from both terrestrial and aquatic species) along with synthetically produced alkaloid compounds having prominent antiviral properties. Previous reviews on this subject have focused on the biological actions of both natural and synthetic alkaloids, but they have not gone into comprehensive detail about their antiviral properties. We reviewed here several antiviral alkaloids that have been described in the literature in different investigational environments i.e. (in-vivo, in-ovo, in-vitro, and in-silico), and found that these alkaloid compounds have significant antiviral properties against several infectious viruses. These alkaloids repressed and targeted various important stages of viral infection at non-toxic doses while some of the alkaloids reported here also exhibited comparable inhibitory activities to commercially used drugs. Overall, these anti-viral effects of alkaloids point to a high degree of specificity, implying that they could serve as effective and safe antiviral medicines if further pursued in medicinal and pharmacological investigations.
Collapse
Affiliation(s)
- Shah Faisal
- Department of Chemistry, Islamia College University Peshawar, Peshawar, 25120, Pakistan
| | - Syed Lal Badshah
- Department of Chemistry, Islamia College University Peshawar, Peshawar, 25120, Pakistan.
| | - Bibi Kubra
- Department of Chemistry, Islamia College University Peshawar, Peshawar, 25120, Pakistan
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
12
|
Rani J, Bhargav A, Khan FI, Ramachandran S, Lai D, Bajpai U. In silico prediction of natural compounds as potential multi-target inhibitors of structural proteins of SARS-CoV-2. J Biomol Struct Dyn 2022; 40:12118-12134. [PMID: 34486935 PMCID: PMC8425474 DOI: 10.1080/07391102.2021.1968497] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a colossal loss to human health and lives and has deeply impacted socio-economic growth. Remarkable efforts have been made by the scientific community in containing the virus by successful development of vaccines and diagnostic kits. Initiatives towards drug repurposing and discovery have also been undertaken. In this study, we compiled the known natural anti-viral compounds using text mining of the literature and examined them against four major structural proteins of SARS-CoV-2, namely, spike (S) protein, nucleocapsid (N) protein, membrane (M) protein and envelope (E) protein. Following computational approaches, we identified fangchinoline and versicolactone C as the compounds to exhibit strong binding to the target proteins and causing structural deformation of three structural proteins (N, S and M). We recommend the inhibitory effects of these compounds from our study should be experimentally validated against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jyoti Rani
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, New Delhi, India,G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research – Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Anasuya Bhargav
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research – Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Faez Iqbal Khan
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Srinivasan Ramachandran
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research – Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,Srinivasan Ramchandran ;
| | - Dakun Lai
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China,Dakun Lai
| | - Urmi Bajpai
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, New Delhi, India,CONTACT Urmi Bajpai ;
| |
Collapse
|
13
|
Sharma D, Sharma N, Manchanda N, Prasad SK, Sharma PC, Thakur VK, Rahman MM, Dhobi M. Bioactivity and In Silico Studies of Isoquinoline and Related Alkaloids as Promising Antiviral Agents: An Insight. Biomolecules 2022; 13:17. [PMID: 36671402 PMCID: PMC9856122 DOI: 10.3390/biom13010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Viruses are widely recognized as the primary cause of infectious diseases around the world. The ongoing global pandemic due to the emergence of SARS-CoV-2 further added fuel to the fire. The development of therapeutics becomes very difficult as viruses can mutate their genome to become more complex and resistant. Medicinal plants and phytocompounds could be alternative options. Isoquinoline and their related alkaloids are naturally occurring compounds that interfere with multiple pathways including nuclear factor-κB, mitogen-activated protein kinase/extracellular-signal-regulated kinase, and inhibition of Ca2+-mediated fusion. These pathways play a crucial role in viral replication. Thus, the major goal of this study is to comprehend the function of various isoquinoline and related alkaloids in viral infections by examining their potential mechanisms of action, structure-activity relationships (SAR), in silico (particularly for SARS-CoV-2), in vitro and in vivo studies. The current advancements in isoquinoline and related alkaloids as discussed in the present review could facilitate an in-depth understanding of their role in the drug discovery process.
Collapse
Affiliation(s)
- Divya Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Neetika Sharma
- Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Namish Manchanda
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Satyendra K. Prasad
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Prabodh Chander Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre, Scotland’s Rural College (SRUC), Kings Buildings, 11 West Mains Road, Edinburgh EH9 3JG, UK
- School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, India
| | - M. Mukhlesur Rahman
- Pharmaceutical and Natural Products Chemistry, School of Health, Sports and Bioscience, University of East London, Stratford Campus, London E15 4LZ, UK
| | - Mahaveer Dhobi
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| |
Collapse
|
14
|
Wang M, Zhang XM, Fu X, Zhang P, Hu WJ, Yang BY, Kuang HX. Alkaloids in genus stephania (Menispermaceae): A comprehensive review of its ethnopharmacology, phytochemistry, pharmacology and toxicology. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115248. [PMID: 35430287 DOI: 10.1016/j.jep.2022.115248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/17/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Approximately 60 species of the genus Stephania (Menispermaceae) are distributed worldwide. Among these, 39 species are located in South and Southwest China; in particular, these plants are rich in alkaloids and were used in traditional Chinese medicine (TCM) against numerous ailments. AIM OF THIS REVIEW The purpose of this study was to provide organized information on the ethnopharmacological uses as well as the phytochemical, pharmacological, and toxicological evaluation of the alkaloids derived from plant species included in the genus Stephania. In addition, we aimed to provide comprehensive basic knowledge on the medicinal properties of these plants and establish meaningful guidelines for further research. MATERIALS AND METHODS Information related to the Stephania genus was collected from scientific databases, such as Web of Science, PubMed, Baidu Scholar, and China Academic Journals (CNKI), within the last 20 years on phytochemistry, pharmacology, and toxicology of the plants in genus Stephania. Furthermore, information was obtained from the Pharmacopoeia of the People's Republic of China. Chinese Pharmacopoeia and Flora of China. RESULTS Plant species belonging to the genus Stephania have been mentioned as traditional remedies and various alkaloidal compounds have been identified and isolated, including aporphine, proaporphine, morphinane, hasubanane, protoberberine, benzylisoquinoline, and bisbenzylisoquinoline and among others. The isolated alkaloidal compounds reportedly exhibited promising pharmacological properties, such as antimicrobial, antiviral, antitumor, antioxidant, antihyperglycemic, anti-inflammatory, antinociceptive, anti-multidrug resistance, neuroprotective, and cardioprotective activities. CONCLUSIONS The genus Stephania is widely used in TCM. The ethnopharmacological uses, phytochemistry, and pharmacology of the Stephania sp. Described in this review demonstrated that these plants contain numerous alkaloids and active constituents and display myriad pharmacological activities. Typically, research on the plants' pharmacological activity focuses on parts of the plants and the associated compounds. However, many Stephania species have rarely been studied, and the ethnomedicinal potential of those discovered has not been scientifically evaluated and needs to be further elucidated. Furthermore, quality control and toxicology studies are warranted in the future.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Xian-Mei Zhang
- Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, 276006, China.
| | - Xin Fu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Peng Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Wen-Jing Hu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Bing-You Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Hai-Xue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
15
|
Dong S, Yu R, Wang X, Chen B, Si F, Zhou J, Xie C, Li Z, Zhang D. Bis-Benzylisoquinoline Alkaloids Inhibit Porcine Epidemic Diarrhea Virus In Vitro and In Vivo. Viruses 2022; 14:v14061231. [PMID: 35746702 PMCID: PMC9228057 DOI: 10.3390/v14061231] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) belongs to the genus Alphacoronavirus of the family Coronaviridae that causes severe diarrhea and high mortality in neonatal suckling piglets. Currently, there is no effective medication against this pathogen. Cepharanthine (CEP), tetrandrine (TET), and fangchinoline (FAN) are natural bis-benzylisoquinoline alkaloids with anti-inflammatory, antitumor, and antiviral properties. Here, we first found that CEP, TET, and FAN had anti-PEDV activity with IC50 values of 2.53, 3.50, and 6.69 μM, respectively. The compounds could block all the processes of viral cycles, but early application of the compounds before or during virus infection was advantageous over application at a late stage of virus replication. FAN performed inhibitory function more efficiently through interfering with the virus entry and attachment processes or through attenuating the virus directly. CEP had a more notable effect on virus entry. With the highest SI index of 11.8 among the three compounds, CEP was chosen to carry out animal experiments. CEP in a safe dosage of 11.1 mg/kg of body weight could reduce viral load and pathological change of piglet intestinal tracts caused by PEDV field strain challenge, indicating that CEP efficiently inhibited PEDV infection in vivo. All of these results demonstrated that the compounds of bis-benzylisoquinoline alkaloids could inhibit PEDV proliferation efficiently and had the potential of being developed for PED prevention and treatment.
Collapse
Affiliation(s)
- Shijuan Dong
- Institute of Animal Husbandry and Veterinary Science, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai Academy of Agricultural Sciences (SAAS), Shanghai 201106, China; (S.D.); (R.Y.); (B.C.); (F.S.); (C.X.)
| | - Ruisong Yu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai Academy of Agricultural Sciences (SAAS), Shanghai 201106, China; (S.D.); (R.Y.); (B.C.); (F.S.); (C.X.)
| | - Xiaoting Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology (ECUST), Shanghai 200237, China;
| | - Bingqing Chen
- Institute of Animal Husbandry and Veterinary Science, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai Academy of Agricultural Sciences (SAAS), Shanghai 201106, China; (S.D.); (R.Y.); (B.C.); (F.S.); (C.X.)
| | - Fusheng Si
- Institute of Animal Husbandry and Veterinary Science, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai Academy of Agricultural Sciences (SAAS), Shanghai 201106, China; (S.D.); (R.Y.); (B.C.); (F.S.); (C.X.)
| | - Jiaming Zhou
- Shanghai Jinshan District Animal Center of Disease Control, Shanghai 201540, China;
| | - Chunfang Xie
- Institute of Animal Husbandry and Veterinary Science, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai Academy of Agricultural Sciences (SAAS), Shanghai 201106, China; (S.D.); (R.Y.); (B.C.); (F.S.); (C.X.)
| | - Zhen Li
- Institute of Animal Husbandry and Veterinary Science, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai Academy of Agricultural Sciences (SAAS), Shanghai 201106, China; (S.D.); (R.Y.); (B.C.); (F.S.); (C.X.)
- Correspondence: (Z.L.); (D.Z.); Tel.: +86-21-62206391 (Z.L.); +86-21-64252324 (D.Z.)
| | - Daojing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology (ECUST), Shanghai 200237, China;
- Correspondence: (Z.L.); (D.Z.); Tel.: +86-21-62206391 (Z.L.); +86-21-64252324 (D.Z.)
| |
Collapse
|
16
|
Ebob OT, Babiaka SB, Ntie-Kang F. Natural Products as Potential Lead Compounds for Drug Discovery Against SARS-CoV-2. NATURAL PRODUCTS AND BIOPROSPECTING 2021; 11:611-628. [PMID: 34515981 PMCID: PMC8435765 DOI: 10.1007/s13659-021-00317-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/01/2021] [Indexed: 05/09/2023]
Abstract
For the past 2 years, the coronavirus responsible for the COVID-19 infection has become a world pandemic, ruining the lives and economies of several nations in the world. This has scaled up research on the virus and the resulting infection with the goal of developing new vaccines and therapies. Natural products are known to be a rich source of lead compounds for drug discovery, including against infectious diseases caused by microbes (viruses, bacteria and fungi). In this review article, we conducted a literature survey aimed at identifying natural products with inhibitory concentrations against the coronaviruses or their target proteins, which lie below 10 µM. This led to the identification of 42 compounds belonging to the alkaloid, flavonoid, terpenoid, phenolic, xanthone and saponin classes. The cut off concentration of 10 µM was to limit the study to the most potent chemical entities, which could be developed into therapies against the viral infection to make a contribution towards limiting the spread of the disease.
Collapse
Affiliation(s)
- Oyere Tanyi Ebob
- Department of Chemistry, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon
| | - Smith B. Babiaka
- Department of Chemistry, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon
- Institute for Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| |
Collapse
|
17
|
Zhou L, Zhou J, Chen T, Chi X, Liu X, Pan S, Chen W, Wu T, Lin T, Zhang X, Li YP, Yang W. Identification of Ascomycin against Zika virus infection through screening of natural product library. Antiviral Res 2021; 196:105210. [PMID: 34801589 DOI: 10.1016/j.antiviral.2021.105210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/27/2021] [Accepted: 11/16/2021] [Indexed: 10/19/2022]
Abstract
Zika virus (ZIKV) infection could lead to Guillain-Barré syndrome in adults and microcephaly in the newborns from infected pregnant women. To date, there is no specific drug for the treatment of ZIKV infection. In this study, we sought to screen inhibitors against ZIKV infection from a natural product library. A ZIKV replicon was used to screen a library containing 1680 natural compounds. We explored the antiviral mechanism of the compound candidate in vitro and in vivo infection models. Ascomycin, a macrolide from Streptomyces hygroscopicus, was identified with inhibitory effect against ZIKV in Vero cells (IC50 = 0.11 μM), hepatoma cell Huh7 (IC50 = 0.38 μM), and glioblastoma cell SNB-19 (IC50 = 0.06 μM), far below the cytotoxic concentrations. Mechanistic study revealed that Ascomycin suppressed ZIKV RNA replication step during the life cycle and the regulation of calcineurin-NFAT pathway maybe involved in this inhibitory effect, independent of innate immunity activation. Moreover, we found that Ascomycin also inhibited the infection of other Flaviviridae members, such as hepatitis C virus and dengue virus. Ascomycin reduced ZIKV load in blood by up to 3500-fold in A129 mice. Meanwhile, the infection in the mice brain was undetectable by immunohistochemistry staining. Together, these findings reveal a critical role of Ascomycin in the inhibition of ZIKV and related viruses, facilitating the development of novel antiviral agents.
Collapse
Affiliation(s)
- Liang Zhou
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Jia Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Tongling Chen
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaojing Chi
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Xiuying Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Shengnan Pan
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Wenfang Chen
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Tiantian Wu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tianli Lin
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Xinhui Zhang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China
| | - Yi-Ping Li
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Wei Yang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100176, China.
| |
Collapse
|
18
|
Yun C, Lee HJ, Lee CJ. Small Molecule Drug Candidates for Managing the Clinical Symptoms of COVID-19: a Narrative Review. Biomol Ther (Seoul) 2021; 29:571-581. [PMID: 34615772 PMCID: PMC8551738 DOI: 10.4062/biomolther.2021.134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
Towards the end of 2019, an atypical acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was identified in Wuhan, China and subsequently named Coronavirus disease 2019 (COVID-19). The rapid dissemination of COVID-19 has provoked a global crisis in public health. COVID-19 has been reported to cause sepsis, severe infections in the respiratory tract, multiple organ failure, and pulmonary fibrosis, all of which might induce mortality. Although several vaccines for COVID-19 are currently being administered worldwide, the COVID-19 pandemic is not yet effectively under control. Therefore, novel therapeutic agents to eradicate the cause of the disease and/or manage the clinical symptoms of COVID-19 should be developed to effectively regulate the current pandemic. In this review, we discuss the possibility of managing the clinical symptoms of COVID-19 using natural products derived from medicinal plants used for controlling pulmonary inflammatory diseases in folk medicine. Diverse natural products have been reported to exert potential antiviral effects in vitro by affecting viral replication, entry into host cells, assembly in host cells, and release. However, the in vivo antiviral effects and clinical antiviral efficacies of these natural products against SARS-CoV-2 have not been successfully proven to date. Thus, these properties need to be elucidated through further investigations, including randomized clinical trials, in order to develop optimal and ideal therapeutic candidates for COVID-19.
Collapse
Affiliation(s)
- Chawon Yun
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyun Jae Lee
- Smith Liberal Arts College and Department of Addiction Science, Graduate School, Sahmyook University, Seoul 01795, Republic of Korea
| | - Choong Jae Lee
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
19
|
Plant Alkaloids Inhibit Membrane Fusion Mediated by Calcium and Fragments of MERS-CoV and SARS-CoV/SARS-CoV-2 Fusion Peptides. Biomedicines 2021; 9:biomedicines9101434. [PMID: 34680551 PMCID: PMC8533497 DOI: 10.3390/biomedicines9101434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/27/2022] Open
Abstract
To rationalize the antiviral actions of plant alkaloids, the ability of 20 compounds to inhibit calcium-mediated fusion of lipid vesicles composed of phosphatidylglycerol and cholesterol was investigated using the calcein release assay and dynamic light scattering. Piperine, tabersonine, hordenine, lupinine, quinine, and 3-isobutyl-1-methylxanthine demonstrated the most potent effects (inhibition index greater than 50%). The introduction of phosphatidylcholine into the phosphatidylglycerol/cholesterol mixture led to significant changes in quinine, hordenine, and 3-isobutyl-1-methylxanthine efficiency. Comparison of the fusion inhibitory ability of the tested alkaloids, and the results of the measurements of alkaloid-induced alterations in the physical properties of model membranes indicated a potent relationship between a decrease in the cooperativity of the phase transition of lipids and the ability of alkaloids to prevent calcium-mediated vesicle fusion. In order to use this knowledge to combat the novel coronavirus pandemic, the ability of the most effective compounds to suppress membrane fusion induced by fragments of MERS-CoV and SARS-CoV/SARS-CoV-2 fusion peptides was studied using the calcein release assay and confocal fluorescence microscopy. Piperine was shown to inhibit vesicle fusion mediated by both coronavirus peptides. Moreover, piperine was shown to significantly reduce the titer of SARS-CoV2 progeny in vitro in Vero cells when used in non-toxic concentrations.
Collapse
|
20
|
da Silva PG, Fonseca AH, Ribeiro MP, Silva TD, Grael CFF, Pena LJ, Silva TMS, Oliveira EDJ. Bisbenzylisoquinoline Alkaloids of Cissampelos Sympodialis With in Vitro Antiviral Activity Against Zika Virus. Front Pharmacol 2021; 12:743541. [PMID: 34603056 PMCID: PMC8482128 DOI: 10.3389/fphar.2021.743541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
In search of new antiviral compounds against Zika virus we conducted a bioassay-guided fractionation of bisbenzyilisoquinoline alkaloids isolated from Cissampelos sympodialis (Menispermaceae), a medicinal plant species endemic to Brazil. Six subfractions were obtained from a tertiary alkaloidal fraction of the rhizomes (TAFrz) using preparative high-performance liquid chromatography. All the subfractions were tested against Zika virus-infected Vero cells as the cellular model to evaluate cytotoxicity and antiviral effective concentrations. The results showed that three of the six TAFrz subfractions tested were active. The most active ones were the subfraction 6 (that consisted of the alkaloids methylwarifteine and warifteine present as a mixture at a ratio of 8.8:1.2 respectively) and the subfraction 5, that was later identified as warifteine, the major tertiary alkaloid of this species. Warifteine was able to significantly reduce virus titer in Zika virus-infected Vero cells with an IC50 of 2.2 μg/ml and this effect was selective (selectivity index, SI = 68.3). Subfraction 6 had an IC50 = 3.5 μg/ml and was more cytotoxic than pure warifteine, with SI = 6.14. Fraction 5 and fraction 6 were more potent in decreasing the viral titer of Zika virus-infected Vero cells than 6-methylmercaptopurine riboside (IC50 = 24.5 μg/ml and SI = 11.9), a mercaptopurine riboside with ZIKV antiviral activity used as a positive control. Our data demonstrate that alkaloids of the bisbenzylisoquinoline type may be explored as new antiviral agents or as an useful pharmacophore for investigating ZIKV antiviral activity.
Collapse
Affiliation(s)
| | - Aventino H Fonseca
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Malu P Ribeiro
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Taizia D Silva
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Cristiane F F Grael
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Lindomar J Pena
- Department of Virology, Aggeu Magalhães Institute (IAM), Fiocruz, Recife, Brazil
| | - Tania M S Silva
- Laboratório de Bioprospecção Fitoquímica, Department of Chemistry, Federal Rural University of Pernambuco, Recife, Brazil
| | | |
Collapse
|
21
|
Abstract
Andrographolide is a labdane diterpenoid extracted and purified from the aerial parts of plants belonging to genus Andrographis (Acanthaceae). The research has shown the plant based compound is low cytotoxic, having antimicrobial, anti-cancer, antiviral and anti-parasitic effects. Andrographolide both prevent spread as well as transmission of virus to neighboring cells by interfering with different cell signaling pathways. In addition to its medicinal value, plant has been found having nutritional value. Therefore being cost effective, easy availability and having nutritional value as a natural supplement, can be used to improve the quality of life in countries having low standard of living. Due to the limited number of effective vaccines, the plant-based antiviral drugs have provided considerable hope for fighting against the viral infections. The plant-derived compound when produced in large quantities is cost effective with low cytotoxic effects. However, much deep insight research at the molecular level is needed to develop the molecules against the viral infection. This paper aims to highlight the antiviral role of Andrographolide that can made significant contributions toward the improvement of human health and will also summarize the current status and future strategies concerning the therapeutic applications of Andrographolide to combat different viral disease in humans.
Collapse
|
22
|
Bibi S, Sarfraz A, Mustafa G, Ahmad Z, Zeb MA, Wang YB, Khan T, Khan MS, Kamal MA, Yu H. Impact of Traditional Plants and their Secondary Metabolites in the Discovery of COVID-19 Treatment. Curr Pharm Des 2021; 27:1123-1143. [PMID: 33213320 DOI: 10.2174/1381612826666201118103416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/12/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Coronavirus Disease-2019 belongs to the family of viruses which cause serious pneumonia along with fever, breathing issues and infection of lungs, and was first reported in China and later spread worldwide. OBJECTIVE Several studies and clinical trials have been conducted to identify potential drugs and vaccines for Coronavirus Disease-2019. The present study listed natural secondary metabolites identified from plant sources with antiviral properties and could be a safer and tolerable treatment for Coronavirus Disease-2019. METHODS A comprehensive search on the reported studies was conducted using different search engines such as Google Scholar, SciFinder, Sciencedirect, Medline PubMed, and Scopus for the collection of research articles based on plant-derived secondary metabolites, herbal extracts, and traditional medicine for coronavirus infections. RESULTS Status of COVID-19 worldwide and information of important molecular targets involved in COVID- 19 are described, and through literature search, it is highlighted that numerous plant species and their extracts possess antiviral properties and are studied with respect to coronavirus treatments. Chemical information, plant source, test system type with a mechanism of action for each secondary metabolite are also mentioned in this review paper. CONCLUSION The present review has listed plants that have presented antiviral potential in the previous coronavirus pandemics and their secondary metabolites, which could be significant for the development of novel and a safer drug which could prevent and cure coronavirus infection worldwide.
Collapse
Affiliation(s)
- Shabana Bibi
- Yunnan Herbal Laboratory, College of Ecology and Environment, Institute of Herbal Biotic Resource, Yunnan University, Kunming 650504, Yunnan, China
| | - Ayesha Sarfraz
- Department of Biosciences, Faculty of Sciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Ghazala Mustafa
- Department of Plant Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Zeeshan Ahmad
- Kohsar Homeopathic Medical College, Rawalpindi, Pakistan
| | - Muhammad A Zeb
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, China
| | - Yuan-Bing Wang
- Yunnan Herbal Laboratory, College of Ecology and Environment, Institute of Herbal Biotic Resource, Yunnan University, Kunming 650504, Yunnan, China
| | - Tahir Khan
- Yunnan Herbal Laboratory, College of Ecology and Environment, Institute of Herbal Biotic Resource, Yunnan University, Kunming 650504, Yunnan, China
| | - Muhammad S Khan
- Department of Biosciences, Faculty of Sciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Mohammad A Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hong Yu
- Yunnan Herbal Laboratory, College of Ecology and Environment, Institute of Herbal Biotic Resource, Yunnan University, Kunming 650504, Yunnan, China
| |
Collapse
|
23
|
Wang Q, Tang B, Cao M. Synthesis, characterization, and fungicidal activity of novel Fangchinoline derivatives. Bioorg Med Chem 2020; 28:115778. [DOI: 10.1016/j.bmc.2020.115778] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 01/04/2023]
|
24
|
Monticolo F, Palomba E, De Santis R, Assentato L, Triscino V, Langella MC, Lanzotti V, Chiusano ML. anti-HCoV: A web resource to collect natural compounds against human coronaviruses. Trends Food Sci Technol 2020; 106:1-11. [PMID: 32982062 PMCID: PMC7505852 DOI: 10.1016/j.tifs.2020.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/29/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND A novel coronavirus, the SARS-CoV2, was revealed to be the cause of COVID19, the pandemic disease that already provoked more than 555.324 deaths in the world (July 10, 2020). No vaccine treatment has been defined against SARS-CoV2 or other human coronaviruses (HCoVs), including those causing epidemic infections, neither appropriate strategies for prevention and care are yet officially suggested. SCOPE AND APPROACH We reviewed scientific literature on natural compounds that were defined as potentially effective against human coronaviruses. Our desk research identified non-chemically modified natural compounds that were shown (in vitro) and/or predicted (in silico) to act against one or more phases of human coronaviruses cell cycle.We selected all available information, merged and annotated the data to define a comprehensive list of natural compounds, describing their chemical classification, the source, the action, the specific target in the viral infection. Our aim was to collect possible compounds for prevention and care against human coronaviruses. KEY FINDINGS AND CONCLUSIONS The definition of appropriate interventions against viral diseases need a comprehensive view on the infection dynamics and on necessary treatments. Viral targeting compounds to be exploited in food sciences could be of relevant interest to this aim.We collected 174 natural compounds showing effects against human infecting coronaviruses, providing a curated annotation on actions and targets.The data are available in anti-HCoV, a web accessible resource to be exploited for testing and in vivo trials. The website is here launched to favour a community based cooperative effort to call for contribution and expand the collection. To be ready to fight.
Collapse
Affiliation(s)
- Francesco Monticolo
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy
| | - Emanuela Palomba
- Department of Research Infrastructures for marine biological resources (RIMAR), Stazione Zoologica "Anton Dohrn", Villa Comunale 80121 - Napoli, Italy
| | - Rosa De Santis
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy
| | - Lorenzo Assentato
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy
| | - Viviana Triscino
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy
| | - Maria Chiara Langella
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy
| | - Virginia Lanzotti
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy
| | - Maria Luisa Chiusano
- Department of Agricultural Sciences, Università degli studi di Napoli Federico II, Reggia di Portici - Via Università, 100 - 80055, Portici (NA), Italy.,Department of Research Infrastructures for marine biological resources (RIMAR), Stazione Zoologica "Anton Dohrn", Villa Comunale 80121 - Napoli, Italy
| |
Collapse
|
25
|
Alkaloids: Therapeutic Potential against Human Coronaviruses. Molecules 2020; 25:molecules25235496. [PMID: 33255253 PMCID: PMC7727683 DOI: 10.3390/molecules25235496] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Alkaloids are a class of natural products known to have wide pharmacological activity and have great potential for the development of new drugs to treat a wide array of pathologies. Some alkaloids have antiviral activity and/or have been used as prototypes in the development of synthetic antiviral drugs. In this study, eleven anti-coronavirus alkaloids were identified from the scientific literature and their potential therapeutic value against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is discussed. In this study, in silico studies showed an affinity of the alkaloids for binding to the receptor-binding domain of the SARS-CoV-2 spike protein, putatively preventing it from binding to the host cell. Lastly, several mechanisms for the known anti-coronavirus activity of alkaloids were discussed, showing that the alkaloids are interesting compounds with potential use as bioactive agents against SARS-CoV-2.
Collapse
|
26
|
Garg S, Roy A. In silico analysis of selected alkaloids against main protease (M pro) of SARS-CoV-2. Chem Biol Interact 2020; 332:109309. [PMID: 33181114 PMCID: PMC7649659 DOI: 10.1016/j.cbi.2020.109309] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/04/2020] [Indexed: 12/26/2022]
Abstract
In the present situation, COVID-19 has become the global health concern due to its high contagious nature. It initially appeared in December 2019 in Wuhan, China and now affected more than 190 countries. As of now preventive measures are the sole solution to stop this disease for further transmission from person to person transmissions as there is no effective treatment or vaccine available to date. Research and development of new molecule is a laborious process; therefore, drug repurposing can be an alternative solution that involves the identification of potential compounds from the already available data. Alkaloids are potential source of therapeutic agents which might be able to treat novel COVID-19. Therefore, in the present study, twenty potential alkaloid molecules that possess antiviral activity against different viral diseases have taken into consideration and scrutinized using Lipinski's rule. Then out of twenty compounds seventeen were further selected for docking study. Docking study was performed using Autodock software and the best four molecule which provides maximum negative binding energy was selected for further analysis. Two alkaloids namely thalimonine and sophaline D showed potential activity to inhibit the Mpro but to confirm the claim further in-vitro studies are required.
Collapse
Affiliation(s)
- Saksham Garg
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Arpita Roy
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India; Department of Biotechnology, Delhi Technological University, Delhi, India.
| |
Collapse
|
27
|
da Silva PG, Fonseca AH, Ribeiro MP, Silva TD, Grael CFF, Pena LJ, Silva TMS, Oliveira EDJ. Bisbenzylisoquinoline alkaloids of Cissampelos sympodialis with antiviral activity against dengue virus. Nat Prod Res 2020; 35:6129-6133. [PMID: 32990038 DOI: 10.1080/14786419.2020.1827404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
A number of bisbenzyilisoquinoline alkaloids have been previously isolated from Cissampelos sympodialis (Menispermaceae). The tertiary alkaloid fraction of the rhizomes (TAFrz) was prepared and the major alkaloid warifteine was isolated. Five TAFrz subfractions in addition to warifteine were tested against Dengue virus (DENV). We then used an epithelial (Vero) cell line to evaluate the cytotoxicity and effective concentrations of the samples against DENV. All TAFrz subfractions were active, but subfraction 6 (a mixture of the alkaloids methylwarifteine and warifteine) in particular showed a promising antiviral effect against DENV-2 with an IC50 of 2.00 µg/mL and a selectivity index (SI) of 10.74. Warifteine was the second most active sample and had an IC50 of 8.13 µg/mL and SI = 10.94. The antiviral activity of the samples compared favorably with that of 6-methylmercaptopurine riboside (IC50 = 7.31 µg/mL and SI = 11.8). These results suggest that bisbenzylisoquinoline alkaloids may prove interesting leading antiviral compounds.
Collapse
Affiliation(s)
- Poliana G da Silva
- Department of Virology, Aggeu Magalhães Institute (IAM), Fiocruz, Pernambuco, Recife, PE, Brazil
| | - Aventino H Fonseca
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| | - Malu P Ribeiro
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| | - Taizia D Silva
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| | - Cristiane F F Grael
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| | - Lindomar J Pena
- Department of Virology, Aggeu Magalhães Institute (IAM), Fiocruz, Pernambuco, Recife, PE, Brazil
| | - Tania M S Silva
- Laboratório de Bioprospecção Fitoquímica, Departamento de Química, Universidade Federal Rural de Pernambuco, Recife, PE, Brazil
| | - Eduardo de J Oliveira
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| |
Collapse
|
28
|
Hazafa A, ur-Rahman K, Haq IU, Jahan N, Mumtaz M, Farman M, Naeem H, Abbas F, Naeem M, Sadiqa S, Bano S. The broad-spectrum antiviral recommendations for drug discovery against COVID-19. Drug Metab Rev 2020; 52:408-424. [PMID: 32546018 PMCID: PMC7309307 DOI: 10.1080/03602532.2020.1770782] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Despite to outbreaks of highly pathogenic beta and alpha coronaviruses including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and human coronavirus, the newly emerged 2019 coronavirus (COVID-19) is considered as a lethal zoonotic virus due to its deadly respiratory syndrome and high mortality rate among the human. Globally, more than 3,517,345 cases have been confirmed with 243,401 deaths due to Acute Respiratory Distress Syndrome (ARDS) caused by COVID-19. The antiviral drug discovery activity is required to control the persistence of COVID-19 circulation and the potential of the future emergence of coronavirus. However, the present review aims to highlight the important antiviral approaches, including interferons, ribavirin, mycophenolic acids, ritonavir, lopinavir, inhibitors, and monoclonal antibodies (mAbs) to provoke the nonstructural proteins and deactivate the structural and essential host elements of the virus to control and treat the infection of COVID-19 by inhibiting the viral entry, viral RNA replication and suppressing the viral protein expression. Moreover, the present review investigates the epidemiology, diagnosis, structure, and replication of COVID-19 for better understanding. It is recommended that these proteases, inhibitors, and antibodies could be a good therapeutic option in drug discovery to control the newly emerged coronavirus.HighlightsCOVID-19 has more than 79.5% identical sequence to SARS-CoV and a 96% identical sequence of the whole genome of bat coronaviruses.Acute respiratory distress syndrome (ARDS), renal failure, and septic shock are the possible clinical symptoms associated with COVID-19.Different antivirals, including interferons, ribavirin, lopinavir, and monoclonal antibodies (mAbs) could be the potent therapeutic agents against COVID-19.The initial clinical trials on hydroquinone in combination with azithromycin showed an admirable result in the reduction of COVID-19.The overexpression of inflammation response, cytokine dysregulation, and induction of apoptosis could be an well-organized factors to reduce the pathogenicity of COVID-19.
Collapse
Affiliation(s)
- Abu Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Khalil ur-Rahman
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Ikram-ul- Haq
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Nazish Jahan
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Mumtaz
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Farman
- Department of Chemistry, University of Engineering and Technology, Lahore, Pakistan
| | - Huma Naeem
- Department of Computer Science, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Faheem Abbas
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Naeem
- Department of Biochemistry, Faculty of Sciences, Bahauddin Zakariya University, Multan, Pakistan
| | - Sania Sadiqa
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Saira Bano
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
29
|
Jiang Y, Liu M, Liu H, Liu S. A critical review: traditional uses, phytochemistry, pharmacology and toxicology of Stephania tetrandra S. Moore (Fen Fang Ji). PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2020; 19:449-489. [PMID: 32336965 PMCID: PMC7180683 DOI: 10.1007/s11101-020-09673-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/15/2020] [Indexed: 05/05/2023]
Abstract
ABSTRACT Stephania tetrandra S. Moore (S. tetrandra) is distributed widely in tropical and subtropical regions of Asia and Africa. The root of this plant is known in Chinese as "Fen Fang Ji". It is commonly used in traditional Chinese medicine to treat arthralgia caused by rheumatism, wet beriberi, dysuria, eczema and inflamed sores. Although promising reports have been published on the various chemical constituents and activities of S. tetrandra, no review comprehensively summarizes its traditional uses, phytochemistry, pharmacology and toxicology. Therefore, the review aims to provide a critical and comprehensive evaluation of the traditional use, phytochemistry, pharmacological properties, pharmacokinetics and toxicology of S. tetrandra in China, and meaningful guidelines for future investigations.
Collapse
Affiliation(s)
- Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008 China
- Institute of Hospital Pharmacy, Central South University, Changsha, 410008 China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| | - Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008 China
- Institute of Hospital Pharmacy, Central South University, Changsha, 410008 China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| | - Haitao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008 China
- Institute of Hospital Pharmacy, Central South University, Changsha, 410008 China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008 China
- Institute of Hospital Pharmacy, Central South University, Changsha, 410008 China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| |
Collapse
|
30
|
Zhou L, Hong G, Li S, Liu Q, Song F, Zhao J, Yuan J, Tickner J, Xu J. Fangchinoline protects against bone loss in OVX mice via inhibiting osteoclast formation, bone resorption and RANKL-induced signaling. Int J Biol Sci 2020; 16:309-319. [PMID: 31929758 PMCID: PMC6949157 DOI: 10.7150/ijbs.37162] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis is a disease characterized by abnormally increased formation and function of osteoclasts. Anti-RANKL treatment using natural medicine is a potential therapy for osteoporosis. Here, we studied the effect of fangchinoline, which is extracted from the root of Stephania tetrandra S. Moore, on osteoclast formation and function. We found that fangchinoline inhibited osteoclastogenesis at doses of 0.5 and 1 µM. In addition, we also examined the mechanism of the inhibitory effect of fangchinoline on osteoclasts. We found that fangchinoline down regulated NFATc1 activity and expression. However, fangchinoline did not affect IκBα degradation and MAPK pathways. In addition, we also found that fangchinoline could protect against bone loss in OVX mice. Taken together, fangchinoline may be a potential compound for osteoporosis.
Collapse
Affiliation(s)
- Lin Zhou
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia.,Department of Endocrinology, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China
| | - Guoju Hong
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia.,Orthopedic Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shangfu Li
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia.,Department of Spine Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, 510630, P. R. China
| | - Qian Liu
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia.,Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi, 530021, China
| | - Fangming Song
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia.,Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi, 530021, China
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi, 530021, China
| | - Jinbo Yuan
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Jennifer Tickner
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, 6009, Australia
| |
Collapse
|
31
|
Natural Bis-Benzylisoquinoline Alkaloids-Tetrandrine, Fangchinoline, and Cepharanthine, Inhibit Human Coronavirus OC43 Infection of MRC-5 Human Lung Cells. Biomolecules 2019; 9:biom9110696. [PMID: 31690059 PMCID: PMC6921063 DOI: 10.3390/biom9110696] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 12/22/2022] Open
Abstract
Stephania tetrandra and other related species of Menispermaceae are the major sources of the bis-benzylisoquinoline alkaloids tetrandrine (TET), fangchinoline (FAN), and cepharanthine (CEP). Although the pharmacological properties of these compounds include anticancer and anti-inflammatory activities, the antiviral effects of these compounds against human coronavirus (HCoV) remain unclear. Hence, the aims of the current study were to assess the antiviral activities of TET, FAN, and CEP and to elucidate the underlying mechanisms in HCoV-OC43-infected MRC-5 human lung cells. These compounds significantly inhibited virus-induced cell death at the early stage of virus infection. TET, FAN, and CEP treatment dramatically suppressed the replication of HCoV-OC43 as well as inhibited viral S and N protein expression. The virus-induced host response was reduced by compound treatment as compared with the vehicle control. Taken together, these findings demonstrate that TET, FAN, and CEP are potential natural antiviral agents for the prevention and treatment of HCoV-OC43 infection.
Collapse
|
32
|
Abstract
The viral infection and resistance to the existing antiviral drugs are alarming, which is a serious public health concern. Medicinal plants are valuable resources for treatment of viral infections and can be used for the management of infections like herpes simplex virus (HSV), human immunodeficiency virus (HIV), influenza, etc. The antiviral screening of plant extracts should be highly selective, specific, and sensitive for bioactivity guided isolation of the active compounds from the plant extracts. The antiviral screening system should be validated for accuracy, reproducibility, simplicity, and cost effectiveness. This chapter highlights on various aspects for screening and evaluation of antiviral natural components including factors affecting antiviral in vivo studies, host cells, organisms, and culture media followed by different virus-specific assays for antiviral screening of natural products.
Collapse
|
33
|
Broad-spectrum antiviral properties of andrographolide. Arch Virol 2016; 162:611-623. [PMID: 27896563 DOI: 10.1007/s00705-016-3166-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/20/2016] [Indexed: 01/01/2023]
Abstract
Andrographolide, a diterpenoid, is known for its anti-inflammatory effects. It can be isolated from various plants of the genus Andrographis, commonly known as 'creat'. This purified compound has been tested for its anti-inflammatory effects in various stressful conditions, such as ischemia, pyrogenesis, arthritis, hepatic or neural toxicity, carcinoma, and oxidative stress, Apart from its anti-inflammatory effects, andrographolide also exhibits immunomodulatory effects by effectively enhancing cytotoxic T cells, natural killer (NK) cells, phagocytosis, and antibody-dependent cell-mediated cytotoxicity (ADCC). All these properties of andrographolide form the foundation for the use of this miraculous compound to restrain virus replication and virus-induced pathogenesis. The present article covers antiviral properties of andrographolide in variety of viral infections, with the hope of developing of a new highly potent antiviral drug with multiple effects.
Collapse
|
34
|
Law BYK, Mok SWF, Wu AG, Lam CWK, Yu MXY, Wong VKW. New Potential Pharmacological Functions of Chinese Herbal Medicines via Regulation of Autophagy. Molecules 2016; 21:359. [PMID: 26999089 PMCID: PMC6274228 DOI: 10.3390/molecules21030359] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a universal catabolic cellular process for quality control of cytoplasm and maintenance of cellular homeostasis upon nutrient deprivation and environmental stimulus. It involves the lysosomal degradation of cellular components such as misfolded proteins or damaged organelles. Defects in autophagy are implicated in the pathogenesis of diseases including cancers, myopathy, neurodegenerations, infections and cardiovascular diseases. In the recent decade, traditional drugs with new clinical applications are not only commonly found in Western medicines, but also highlighted in Chinese herbal medicines (CHM). For instance, pharmacological studies have revealed that active components or fractions from Chaihu (Radix bupleuri), Hu Zhang (Rhizoma polygoni cuspidati), Donglingcao (Rabdosia rubesens), Hou po (Cortex magnoliae officinalis) and Chuan xiong (Rhizoma chuanxiong) modulate cancers, neurodegeneration and cardiovascular disease via autophagy. These findings shed light on the potential new applications and formulation of CHM decoctions via regulation of autophagy. This article reviews the roles of autophagy in the pharmacological actions of CHM and discusses their new potential clinical applications in various human diseases.
Collapse
Affiliation(s)
- Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Simon Wing Fai Mok
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - An Guo Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Christopher Wai Kei Lam
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Margaret Xin Yi Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
35
|
Zhu H, Chen C, Liu J, Sun B, Wei G, Li Y, Zhang J, Yao G, Luo Z, Xue Y, Zhang Y. Hyperascyrones A-H, polyprenylated spirocyclic acylphloroglucinol derivatives from Hypericum ascyron Linn. PHYTOCHEMISTRY 2015; 115:222-230. [PMID: 25800107 DOI: 10.1016/j.phytochem.2015.02.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/28/2015] [Accepted: 02/05/2015] [Indexed: 06/04/2023]
Abstract
Eight polyprenylated spirocyclic acylphloroglucinol derivatives (PSAPs), hyperascyrones A-H, were isolated from the aerial parts of Hypericum ascyron Linn., together with six known analogs. Their structures were established by spectroscopic analyses including HRESIMS, 1D and 2D NMR, and their absolute configurations were determined by electronic circular dichroism calculations (ECD, Gaussian 09). Structures of previously reported tomoeones C, D, G, and H were revised. Hyperascyrones A-H were evaluated for their cytotoxic and anti-HIV-1 activities, with hyperascyrones C and G exhibiting significant cytotoxicities against HL-60 cell lines with IC50 values of 4.22 and 8.36 μM, respectively. In addition, the chemotaxonomic significance of these compounds was also discussed.
Collapse
Affiliation(s)
- Hucheng Zhu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chunmei Chen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Junjun Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Bin Sun
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Guangzheng Wei
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, People's Republic of China
| | - Jinwen Zhang
- Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Guangmin Yao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zengwei Luo
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yongbo Xue
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|
36
|
Chattopadhyay D, Ojha D, Mondal S, Goswami D. Validation of Antiviral Potential of Herbal Ethnomedicine. EVIDENCE-BASED VALIDATION OF HERBAL MEDICINE 2015. [PMCID: PMC7150199 DOI: 10.1016/b978-0-12-800874-4.00008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Natural products are the basis of treatment since the dawn of human civilization, and modern medicine has gradually developed, over the years, by scientific and observational efforts from traditional medicine. Today most of the synthetic drugs showed adverse and unacceptable side effects, however, impressive bioactivities with reduced toxicities were reported for many botanicals against several chronic or difficult-to-treat diseases. A whole range of viral diseases including human immunodeficiency virus/acquired immunodeficiency syndrome, severe acute respiratory syndrome, Rabies, Dengue, and Herpes need effective drugs. Considerable research has been carried out on the pharmacognosy, chemistry, pharmacology, and therapeutics of traditional medicines of diverse cultures, and many pharmaceutical companies have renewed their strategies for antiviral drug development where no effective drugs or vaccine exist. Thus, phytochemicals with antiviral potentials need to be studied in depth with standardization, chemical isolation, effectivity, molecular mechanism, along with in vivo toxicity and efficacy to reduce cost and time. This review will portray the scientific approaches and methodologies used for the development of antiviral leads from traditional medicines against selected genetically and functionally diverse viral infections.
Collapse
|
37
|
Zhu H, Chen C, Yang J, Li XN, Liu J, Sun B, Huang SX, Li D, Yao G, Luo Z, Li Y, Zhang J, Xue Y, Zhang Y. Bioactive Acylphloroglucinols with Adamantyl Skeleton from Hypericum sampsonii. Org Lett 2014; 16:6322-5. [PMID: 25453445 DOI: 10.1021/ol5030579] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Hucheng Zhu
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunmei Chen
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Yang
- State
Key Laboratory of Phytochemistry and Plant Resources in West China,
Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao-Nian Li
- State
Key Laboratory of Phytochemistry and Plant Resources in West China,
Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
| | - Junjun Liu
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Sun
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng-Xiong Huang
- State
Key Laboratory of Phytochemistry and Plant Resources in West China,
Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
| | - Dongyan Li
- Tongji
Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guangmin Yao
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zengwei Luo
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Li
- State
Key Laboratory of Phytochemistry and Plant Resources in West China,
Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
| | - Jinwen Zhang
- Tongji
Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongbo Xue
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yonghui Zhang
- Hubei
Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation,
School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
38
|
Hu ZX, Xue YB, Bi XB, Zhang JW, Luo ZW, Li XN, Yao GM, Wang JP, Zhang YH. Five new secondary metabolites produced by a marine-associated fungus, Daldinia eschscholzii. Mar Drugs 2014; 12:5563-75. [PMID: 25419997 PMCID: PMC4245545 DOI: 10.3390/md12115563] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 12/17/2022] Open
Abstract
Five new compounds, including a benzopyran ribonic glycoside, daldiniside A (1), two isocoumarin ribonic glycosides, daldinisides B (2) and C (3), and two alkaloids, 1-(3-indolyl)-2R,3-dihydroxypropan-1-one (4) and 3-ethyl-2,5-pyrazinedipropanoic acid (5), along with five known compounds (6–10), were isolated from the EtOAc extract of the marine-associated fungus, Daldinia eschscholzii. Their structures were elucidated by extensive physicochemical and spectroscopic properties, besides comparison with literature data. The absolute configurations of compounds 1–3 were corroborated by chemical transformation, GC analysis and X-ray crystallographic analysis. Meanwhile, the absolute configuration of compound 4 and the planar structure of compound 6 were also determined based on the X-ray diffraction analysis. The cytotoxicity of compounds 1–10, antifungal and anti-HIV activities of compounds 1–5 and the in vitro assay for glucose consumption of compounds 1–3 were done in the anti-diabetic model, whereas none showed obvious activity.
Collapse
Affiliation(s)
- Zheng-Xi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yong-Bo Xue
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiao-Bin Bi
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jin-Wen Zhang
- Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zeng-Wei Luo
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiao-Nian Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China.
| | - Guang-Min Yao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jian-Ping Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yong-Hui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
39
|
|
40
|
Colletotrichumine A, a novel indole–pyrazine alkaloid with an unprecedented C16N3-type skeleton from cultures of Colletotrichum capsici. Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.09.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Desgrouas C, Taudon N, Bun SS, Baghdikian B, Bory S, Parzy D, Ollivier E. Ethnobotany, phytochemistry and pharmacology of Stephania rotunda Lour. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:537-563. [PMID: 24768769 DOI: 10.1016/j.jep.2014.04.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Stephania rotunda Lour. (Menispermaceae) is an important traditional medicinal plant that is grown in Southeast Asia. The stems, leaves, and tubers have been used in the Cambodian, Lao, Indian and Vietnamese folk medicine systems for years to treat a wide range of ailments, including asthma, headache, fever, and diarrhoea. AIM OF THE REVIEW To provide an up-to-date, comprehensive overview and analysis of the ethnobotany, phytochemistry, and pharmacology of Stephania rotunda for its potential benefits in human health, as well as to assess the scientific evidence of traditional use and provide a basis for future research directions. MATERIAL AND METHODS Peer-reviewed articles on Stephania rotunda were acquired via an electronic search of the major scientific databases (Pubmed, Google Scholar, and ScienceDirect). Data were collected from scientific journals, theses, and books. RESULTS The traditional uses of Stephania rotunda were recorded in countries throughout Southeast Asia (Cambodia, Vietnam, Laos, and India). Different parts of Stephania rotunda were used in traditional medicine to treat about twenty health disorders. Phytochemical analyses identified forty alkaloids. The roots primarily contain l-tetrahydropalmatine (l-THP), whereas the tubers contain cepharanthine and xylopinine. Furthermore, the chemical composition differs from one region to another and according to the harvest period. The alkaloids exhibited approximately ten different pharmacological activities. The main pharmacological activities of Stephania rotunda alkaloids are antiplasmodial, anticancer, and immunomodulatory effects. Sinomenine, cepharanthine, and l-stepholidine are the most promising components and have been tested in humans. The pharmacokinetic parameters have been studied for seven compounds, including the three most promising compounds. The toxicity has been evaluated for liriodenine, roemerine, cycleanine, l-tetrahydropalmatine, and oxostephanine. CONCLUSION Stephania rotunda is traditionally used for the treatment of a wide range of ailments. Pharmacological investigations have validated different uses of Stephania rotunda in folk medicine. The present review highlights the three most promising compounds of Stephania rotunda, which could constitute potential leads in various medicinal fields, including malaria and cancer.
Collapse
Affiliation(s)
- Camille Desgrouas
- UMR-MD3, IRBA, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France; UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | | | - Sok-Siya Bun
- UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | - Beatrice Baghdikian
- UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | - Sothavireak Bory
- Faculté de Pharmacie, Université des Sciences de la Santé, no. 73, Monivong Blvd, Daun Penh, Phnom Penh, Cambodia.
| | - Daniel Parzy
- UMR-MD3, IRBA, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | - Evelyne Ollivier
- UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| |
Collapse
|
42
|
Liao Q, Qian Z, Liu R, An L, Chen X. Germacrone inhibits early stages of influenza virus infection. Antiviral Res 2013; 100:578-88. [PMID: 24095670 DOI: 10.1016/j.antiviral.2013.09.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 09/20/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
Abstract
Highly pathogenic influenza viruses pose a serious public health threat to humans. Although vaccines are available, antivirals are needed to efficiently control disease progression and virus transmission due to the emergence of drug-resistant viral strains. In this study, germacrone, which is a major component of the essential oils extracted from Rhizoma Curcuma, was found to inhibit influenza virus replication. Germacrone showed antiviral activity against the H1N1 and H3N2 influenza A viruses and the influenza B virus in a dose-dependent manner. The viral protein expression, RNA synthesis and the production of infectious progeny viruses were decreased both in MDCK and A549 cells treated with germacrone. In a time-of-addition study, germacrone was found to exhibit an inhibitory effect on both the attachment/entry step and the early stages of the viral replication cycle. Germacrone also exhibited an effective protection of mice from lethal infection and reduced the virus titres in the lung. Furthermore, the combination of germacrone and oseltamivir exhibited an additive effect on the inhibition of influenza virus infection, both in vitro and in vivo. Our results suggest that germacrone may have the potential to be developed as a therapeutic agent alone or in combination with other agents for the treatment of influenza virus infection.
Collapse
Affiliation(s)
- Qingjiao Liao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academic of Sciences, Wuhan, Hubei 430071, PR China
| | | | | | | | | |
Collapse
|