1
|
Zhou F, Deng S, Luo Y, Liu Z, Liu C. Research Progress on the Protective Effect of Green Tea Polyphenol (-)-Epigallocatechin-3-Gallate (EGCG) on the Liver. Nutrients 2025; 17:1101. [PMID: 40218859 PMCID: PMC11990830 DOI: 10.3390/nu17071101] [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: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
The liver, as the primary metabolic organ, is susceptible to an array of factors that can harm liver cells and give rise to different liver diseases. Epigallocatechin gallate (EGCG), a natural compound found in green tea, exerts numerous beneficial effects on the human body. Notably, EGCG displays antioxidative, antibacterial, antiviral, anti-inflammatory, and anti-tumor properties. This review specifically highlights the pivotal role of EGCG in liver-related diseases, focusing on viral hepatitis, autoimmune hepatitis, fatty liver disease, and hepatocellular carcinoma. EGCG not only inhibits the entry and replication of hepatitis B and C viruses within hepatocytes, but also mitigates hepatocytic damage caused by hepatitis-induced inflammation. Furthermore, EGCG exhibits significant therapeutic potential against hepatocellular carcinoma. Combinatorial use of EGCG and anti-hepatocellular carcinoma drugs enhances the sensitivity of drug-resistant cancer cells to chemotherapeutic agents, leading to improved therapeutic outcomes. Thus, the combination of EGCG and anti-hepatocellular carcinoma drugs holds promise as an effective approach for treating drug-resistant hepatocellular carcinoma. In conclusion, EGCG possesses hepatoprotective properties against various forms of liver damage and emerges as a potential drug candidate for liver diseases.
Collapse
Affiliation(s)
- Fang Zhou
- School of Chemistry and Environmental Sciences, Xiangnan University, Chenzhou 423000, China;
| | - Sengwen Deng
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China; (S.D.); (C.L.)
| | - Yong Luo
- School of Chemistry and Environmental Sciences, Xiangnan University, Chenzhou 423000, China;
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China;
| | - Changwei Liu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China; (S.D.); (C.L.)
| |
Collapse
|
2
|
Warinhomhoun S, Raiputta J, Rangsee PN, Yang CS, Chueamchaitrakun P. Effect of Ultrasound-Assisted Extraction and Drying Methods on Bioactive Compounds, Phenolic Composition, and Antioxidant Activity of Assam Tea Cultivar ( Camellia sinensis var. assamica) Cultivated in Thailand. Adv Pharmacol Pharm Sci 2024; 2024:5772961. [PMID: 39512303 PMCID: PMC11540878 DOI: 10.1155/2024/5772961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Tea is a rich source of phytochemicals; their composition in tea extracts varies depending on the cultivar, climate, production region, and processing and handling processes. The method of extraction plays a crucial role in determining the biological effects of the bioactive compounds in tea leaves. However, reports on the catechin profiles and antioxidant activities of the extracts obtained from leaves at different stages of maturity are limited. Here, we aimed to evaluate the effect of ultrasound-assisted extraction (UAE) and different drying methods, freeze drying (FD) and spray drying (SD), on the composition of bioactive compounds, phenolic composition, and antioxidant activity of extracts obtained from different part of leaves, top (TT), middle (ML), and mature (MT), of Assam tea cultivar (Camellia sinensis var. assamica) cultivated in Thailand (Thai Assam tea). High-performance liquid chromatography analysis showed that the extracts obtained by UAE with FD from TT leaves (UAEFD-TT) had the highest catechins (341.38 ± 0.11 mg/g extract) and caffeine (93.20 ± 0.36 mg CF/g extract) contents compared with those extracted from ML and MT using the same method as well those obtained by SD. The total phenolic and total flavonoid contents were the highest in UAEFD-TT extracts (456.78 ± 4.31 mg GAE/g extract and 333.98 ± 0.83 mg QE/g extract, respectively). In addition, UAEFD-TT exhibited the highest antioxidant activity; the IC50 values obtained by 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) assays were 1.31 ± 0.02 and 7.51 ± 0.03 μg/mL, respectively. In the ferric-reducing antioxidant power (FRAP) assay, the UAEFD-TT extract demonstrated the highest antioxidant activity (324.54 ± 3.33 μM FeSO4/mg extract). These results suggest that extraction from TT using UAE followed by FD produced the highest amount of antioxidant compounds in Thai Assam tea extracts.
Collapse
Affiliation(s)
- Sakan Warinhomhoun
- College of Oriental Medicine, Rangsit University, Lak Hok 12000, Pathum Thani, Thailand
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Center of Excellent in Marijuana, Hemp and Kratom, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Jiraporn Raiputta
- Tea and Coffee Institute, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Paryn Na Rangsee
- Tea and Coffee Institute, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Chung S. Yang
- Department of Chemical Biology, Rutgers University, 164 Frelinghuysen Road, Piscataway 08854, New Jersey, USA
| | - Piyaporn Chueamchaitrakun
- Tea and Coffee Institute, Mae Fah Luang University, Chiang Rai 57100, Thailand
- School of Agro-Industry, Mae Fah Luang University, Chiang Rai 57100, Thailand
| |
Collapse
|
3
|
Sharma KK, Devi S, Kumar D, Ali Z, Fatma N, Misra R, Kumar G. Role of Natural Products against the Spread of SARS-CoV-2 by Inhibition of ACE-2 Receptor: A Review. Curr Pharm Des 2024; 30:2562-2573. [PMID: 39041269 DOI: 10.2174/0113816128320161240703092622] [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: 03/30/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
A unique extreme acute breathing syndrome emerged in China and spread rapidly globally due to a newly diagnosed human coronavirus and declared a pandemic. COVID-19 was formally named by WHO, and the Global Committee on Taxonomy referred to it as extreme Acute respiratory Syndrome Coronavirus-2 (SARS-CoV-2). Currently there is no efficient method to control the extent of SARS-CoV-2 other than social distancing and hygiene activities. This study aims to present a simple medicinal strategy for combating fatal viral diseases like COVID-19 with minimum effort and intervention. Different Ayurveda medicines (Curcuma longa, green tea, and Piper nigrum) inhibit virus entrance and pathogen transmission while also enhancing immunity. Piperine (1-piperoylpiperidine), as well as curcumin, combine to create an intermolecular complex (π- π) that improves curcumin bioavailability by inhibiting glucuronidation of curcumin in the liver. The receptor- binding domains of the S-protein and also the angiotensin-converting enzyme 2 receptor of the recipient organism are directly occupied by curcumin and catechin, respectively, thereby preventing viruses from entering the cell. As a result, the infection will be tolerated by the animal host.
Collapse
Affiliation(s)
- Krishana Kumar Sharma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad 244001 (UP), India
| | - Shoma Devi
- Department of Zoology, Krishna College of Science & Information Technology, Bijnor 246701 (UP), India
| | - Dharmendra Kumar
- Science Branch, Pt. Deendayal Upadhyay Institute of Archaeology, Archaeological Survey of India, Greater Noida 201013, India
| | - Zeeshan Ali
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad 244001 (UP), India
| | - Nishat Fatma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad 244001 (UP), India
| | - Raghvendra Misra
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad 244001 (UP), India
| | - Gajendra Kumar
- Department of Chemistry, Constituent Government College, MJP Rohilkhand University, Bareilly, Hasanpur, Amroha 244241 (UP), India
| |
Collapse
|
4
|
Ahmad Merza Mohammad T. Combining nano-curcumin with catechin improves COVID-19-infected patient's inflammatory conditions. Hum Immunol 2023; 84:471-483. [PMID: 37331910 PMCID: PMC10239908 DOI: 10.1016/j.humimm.2023.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/30/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023]
Abstract
AIMS A hyperinflammatory condition is brought on by the development of Coronavirus disease 2019 (COVID-19), which is characterized by an elevation of T helper (Th) 17 cells, high levels of pro-inflammatory cytokines, and a depletion of regulatory T (Treg) cells. METHODS In this research, we examined the effect of nano-curcumin and catechin on the TCD4+, TCD8+, Th17, and Treg cells and their associated factors in COVID-19 patients. For this purpose, 160 (50 patients excluded during the study) COVID-19 patients were divided into four groups: placebo, nano-curcumin, catechin, and nano-curcumin + catechin. The frequency of TCD4+, TCD8+, Th17, and Treg cells, the gene expression of transcription factors (STAT3, RORt, and FoxP3) relevant to Th17 and Treg, as well as the serum levels of cytokines (IL-6, IL17, IL1-b, IL-10, and TGF-), were all evaluated intra- and inter-group, before and after treatment, in all groups. RESULTS Our study showed that TCD4 + and TCD8 + cells were significantly higher in the nano-curcumin + catechin group compared to the control group, whereas Th17 was lower than the initial value. Furthermore, compared to the placebo-received group, cytokines and transcription factors associated with Th17 were significantly lower in the nano-curcumin + catechin group. Additionally, combined therapy increased Treg cells and transcription factors compared to the placebo group. CONCLUSION Overall, our results show that combining nano-curcumin with catechin has a more notable impact on the enhancement of TCD4+, TCD8+, and Treg cells, as well as a decrease in Th17 cells and their mediators, suggesting a promising combination therapy in reducing the inflammatory conditions of COVID-19 infected patients.
Collapse
|
5
|
Nabil-Adam A, E. Elnosary M, L. Ashour M, M. Abd El-Moneam N, A. Shreadah M. Flavonoids Biosynthesis in Plants as a Defense Mechanism: Role and Function Concerning Pharmacodynamics and Pharmacokinetic Properties. FLAVONOID METABOLISM - RECENT ADVANCES AND APPLICATIONS IN CROP BREEDING 2023. [DOI: 10.5772/intechopen.108637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Flavonoids are a major class of secondary metabolites that comprises more than 6000 compounds that have been identified. They are biosynthesized via the phenylpropanoid metabolic pathway that involves groups of enzymes such as isomerases, hydroxylases, and reductases that greatly affect the determination of the flavonoid skeleton. For example, transferase enzymes responsible for the modification of sugar result in changes in the physiological activity of the flavonoids and changes in their physical properties, such as solubility, reactivity, and interaction with cellular target molecules, which affect their pharmacodynamics and pharmacokinetic properties. In addition, flavonoids have diverse biological activities such as antioxidants, anticancer, and antiviral in managing Alzheimer’s disease. However, most marine flavonoids are still incompletely discovered because marine flavonoid biosynthesis is produced and possesses unique substitutions that are not commonly found in terrestrial bioactive compounds. The current chapter will illustrate the importance of flavonoids’ role in metabolism and the main difference between marine and terrestrial flavonoids.
Collapse
|
6
|
Sadiea RZ, Sultana S, Chaki BM, Islam T, Dash S, Akter S, Islam MS, Kazi T, Nagata A, Spagnuolo R, Mancina RM, Hossain MG. Phytomedicines to Target Hepatitis B Virus DNA Replication: Current Limitations and Future Approaches. Int J Mol Sci 2022; 23:ijms23031617. [PMID: 35163539 PMCID: PMC8836293 DOI: 10.3390/ijms23031617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023] Open
Abstract
Hepatitis B virus infection (HBV) is one of the most common causes of hepatitis, and may lead to cirrhosis or hepatocellular carcinoma. According to the World Health Organization (WHO), approximately 296 million people worldwide are carriers of the hepatitis B virus. Various nucleos(t)ide analogs, which specifically suppress viral replication, are the main treatment agents for HBV infection. However, the development of drug-resistant HBV strains due to viral genomic mutations in genes encoding the polymerase protein is a major obstacle to HBV treatment. In addition, adverse effects can occur in patients treated with nucleos(t)ide analogs. Thus, alternative anti-HBV drugs of plant origin are being investigated as they exhibit excellent safety profiles and have few or no side effects. In this study, phytomedicines/phytochemicals exerting significant inhibitory effects on HBV by interfering with its replication were reviewed based on different compound groups. In addition, the chemical structures of these compounds were developed. This will facilitate their commercial synthesis and further investigation of the molecular mechanisms underlying their effects. The limitations of compounds previously screened for their anti-HBV effect, as well as future approaches to anti-HBV research, have also been discussed.
Collapse
Affiliation(s)
- Rahila Zannat Sadiea
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (R.Z.S.); (S.S.); (T.I.)
| | - Shahnaj Sultana
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (R.Z.S.); (S.S.); (T.I.)
| | - Bijan Mohon Chaki
- Department of Chemistry (Organic Chemistry Division), Begum Rokeya University, Rangpur 5400, Bangladesh;
| | - Tasnim Islam
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (R.Z.S.); (S.S.); (T.I.)
| | - Sharmy Dash
- Department of Pharmacology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh;
| | - Sharmin Akter
- Department of Physiology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh;
| | - Md Sayeedul Islam
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka 560-0043, Japan;
| | - Taheruzzaman Kazi
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (T.K.); (A.N.)
| | - Abir Nagata
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (T.K.); (A.N.)
| | - Rocco Spagnuolo
- Experimental and Clinical Medicine Department, Magna Graecia University, 88100 Catanzaro, Italy;
| | | | - Md Golzar Hossain
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (R.Z.S.); (S.S.); (T.I.)
- Correspondence:
| |
Collapse
|
7
|
Teixeira AM, Sousa C. A Review on the Biological Activity of Camellia Species. Molecules 2021; 26:molecules26082178. [PMID: 33918918 PMCID: PMC8069326 DOI: 10.3390/molecules26082178] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/19/2021] [Accepted: 04/04/2021] [Indexed: 02/06/2023] Open
Abstract
Medicinal plants have been used since antiquity to cure illnesses and injuries. In the last few decades, natural compounds extracted from plants have garnered the attention of scientists and the Camellia species are no exception. Several species and cultivars are widespread in Asia, namely in China, Japan, Vietnam and India, being also identified in western countries like Portugal. Tea and oil are the most valuable and appreciated Camellia subproducts extracted from Camellia sinensis and Camellia oleifera, respectively. The economic impact of these species has boosted the search for additional information about the Camellia genus. Many studies can be found in the literature reporting the health benefits of several Camellia species, namely C. sinensis, C. oleifera and Camellia japonica. These species have been highlighted as possessing antimicrobial (antibacterial, antifungal, antiviral) and antitumoral activity and as being a huge source of polyphenols such as the catechins. Particularly, epicatechin (EC), epigallocatechin (EGC), epicatechin-3-gallate (ECG), and specially epigallocatechin-3-gallate (EGCG), the major polyphenols of green tea. This paper presents a detailed review of Camellia species’ antioxidant properties and biological activity.
Collapse
Affiliation(s)
- Ana Margarida Teixeira
- LAQV/REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-290 Porto, Portugal;
| | - Clara Sousa
- CBQF—Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
- Correspondence:
| |
Collapse
|
8
|
Kumar G, Kumar D, Singh NP. Therapeutic Approach against 2019-nCoV by Inhibition of ACE-2 Receptor. Drug Res (Stuttg) 2021; 71:213-218. [PMID: 33184809 PMCID: PMC8043666 DOI: 10.1055/a-1275-0228] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
The continued spread of 2019-nCoV has prompted widespread concern around the world. WHO formally named COVID-19 and International Committee on Taxonomy called it Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). Due to this viral attack, the whole world is in lockdown. Presently, there is no effective way to control it, except social distancing and hygienic activity. World class scientists and researchers are trying to make vaccine and discover the medicine against the control and cure to this deadly viral disease. Our aim to presenting this article is kick-off deadly viral disease i.e COVID-19 by an easy way with minimum intervention and effort. Different ayurvedic therapeutic agents (Curcuma Longa L, Green tea and Piper nigrum) inhabit entry of virus in host cell, transmission of pathogen and improve the immunity. Curcumin and piperine (1-piperoylpiperidine) interact to each other and form a π-π intermolecular complex which enhance the bioavailability of curcumin by inhibition of glucuronidation of curcumin in liver. Both the molecules curcumin and catechin get bound directly to receptors binding domain of S-protein and ACE-2 receptors of host cell, due to which these molecules inhibit the entry of virus in host cell i. e. animal survives from being infected.
Collapse
Affiliation(s)
- Gajendra Kumar
- Department of Chemistry, Krishna College, Bijnor, Uttar Pradesh,
India
| | - Dharmendra Kumar
- Science Branch, Archeological Survey of India, Patna Zone, Behar,
India
| | - Netra Pal Singh
- Department of Chemistry, Deen Dayal Upadhyaya Gorakhpur University,
Gorakhpur, Uttar Pradesh, India
| |
Collapse
|
9
|
Bhowmik D, Nandi R, Prakash A, Kumar D. Evaluation of flavonoids as 2019-nCoV cell entry inhibitor through molecular docking and pharmacological analysis. Heliyon 2021; 7:e06515. [PMID: 33748510 PMCID: PMC7955945 DOI: 10.1016/j.heliyon.2021.e06515] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/21/2020] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
The outbreak of Coronavirus Disease 2019 (COVID-19) has been declared as a Public Health Emergency of International Concern (PHEIC) by the World Health Organization (WHO), which is being rapidly spread by the extremely spreadable and pathogenic 2019 novel coronavirus (2019-nCoV), also known as SARS-CoV-2. Pandemic incidence of COVID-19 has created a severe threat to global public health, necessitating the development of effective drugs or inhibitors or therapeutics agents against SARS-CoV-2. Spike protein (S) of the SARS-CoV-2 plays a crucial role in entering viruses into the host cell by binding to angiotensin-converting enzyme 2 (ACE-2), and this specific interaction represents a promising drug target for the identification of potential drugs. This study aimed at the receptor-binding domain of S protein (RBD of nCoV-SP) and the ACE-2 receptor as a promising target for developing drugs against SARS-CoV-2. Over 100 different flavonoids with antioxidant, anti-inflammatory, and antiviral properties from different literatures were taken as a ligand or inhibitor for molecular docking against target protein RBD of nCoV-SP and ACE-2 using PyRX and iGEMDOCK. Top flavonoids based on docking scores were selected for the pharmacokinetic study. Selected flavonoids (hesperidin, naringin, ECGC, and quercetin) showed excellent pharmacokinetics with proper absorption, solubility, permeability, distribution, metabolism, minimal toxicity, and excellent bioavailability. Molecular dynamics simulation studies up to 100 ns exhibited strong binding affinity of selected flavonoids to RBD of nCoV-SP and ACE-2, and the protein-ligand complexes were structurally stable. These identified lead flavonoids may act as potential compounds for developing effective drugs against SARS-CoV-2 by potentially inhibiting virus entry into the host cell.
Collapse
Affiliation(s)
- Deep Bhowmik
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India
| | - Rajat Nandi
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon, 122413, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India
| |
Collapse
|
10
|
Jena AB, Kanungo N, Nayak V, Chainy GBN, Dandapat J. Catechin and curcumin interact with S protein of SARS-CoV2 and ACE2 of human cell membrane: insights from computational studies. Sci Rep 2021; 11:2043. [PMID: 33479401 PMCID: PMC7820253 DOI: 10.1038/s41598-021-81462-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 12/17/2020] [Indexed: 12/28/2022] Open
Abstract
The recent outbreak of the coronavirus (SARS-CoV2) is an unprecedented threat to human health and society across the globe. In this context, development of suitable interventions is the need of the hour. The viral spike protein (S Protein) and the cognate host cell receptor ACE2 can be considered as effective and appropriate targets for interventions. It is evident from the present computational study, that catechin and curcumin, not only exhibit strong binding affinity to viral S Protein and host receptor ACE2 but also to their complex (receptor-binding domain (RBD) of the spike protein of SARS-CoV2 and ACE2; RBD/ACE2-complex). The binding affinity values of catechin and curcumin for the S protein, ACE2 and RBD/ACE2-complex are − 10.5 and − 7.9 kcal/mol; − 8.9 and − 7.8 kcal/mol; and − 9.1 and − 7.6 kcal/mol, respectively. Curcumin directly binds to the receptor binding domain (RBD) of viral S Protein. Molecular simulation study over a period of 100 ns further substantiates that such interaction within RBD site of S Protein occurs during 40–100 ns out of 100 ns simulation trajectory. Contrary to this, catechin binds with amino acid residues present near the RBD site of S Protein and causes fluctuation in the amino acid residues of the RBD and its near proximity. Both catechin and curcumin bind the interface of ‘RBD/ACE2-complex’ and intervene in causing fluctuation of the alpha helices and beta-strands of the protein complex. Protein–protein interaction studies in presence of curcumin or catechin also corroborate the above findings suggesting the efficacy of these two polyphenols in hindering the formation of S Protein-ACE2 complex. In conclusion, this computational study for the first time predicts the possibility of above two polyphenols for therapeutic strategy against SARS-CoV2.
Collapse
Affiliation(s)
- Atala B Jena
- Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar, 751004, Odisha, India
| | - Namrata Kanungo
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, 751004, Odisha, India
| | - Vinayak Nayak
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, 751004, Odisha, India
| | - G B N Chainy
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, 751004, Odisha, India
| | - Jagneshwar Dandapat
- Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar, 751004, Odisha, India. .,Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, 751004, Odisha, India.
| |
Collapse
|
11
|
Kumar G, Kumar D, Singh NP. Therapeutic Approach Against 2019-nCoV by Inhibition of the ACE-2 receptor. Drug Res (Stuttg) 2020. [PMID: 33368058 DOI: 10.1055/a-1337-4462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The continued spread of the 2019 novel coronavirus (2019-nCoV) has prompted global concern. The formal name given to 2019-nCoV by the World Health Organization is COVID-19, while the International Committee on Taxonomy has named it severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Due to this viral attack, nations around the world have issued lockdown restrictions. Presently, there is no effective way to control the spread of 2019-nCoV, except through social distancing and hygienic activities. World-class scientists and researchers are trying to develop vaccines and medicines that will cure this deadly viral disease and control its spread. Our aim in presenting this article is to provide an easy therapeutic approach that effectively combats deadly viral diseases, such as COVID-19, with minimal intervention and effort. Different Ayurvedic therapeutic agents (Curcuma longa L, green tea, and Piper nigrum) inhibit the entry of viruses in the host cell and the transmission of pathogens, while improving immunity. Curcumin and piperine (1-piperoylpiperidine) interact with each other and form a π-π intermolecular complex that enhances the bioavailability of curcumin by inhibition of glucuronidation of curcumin in the liver. Two molecules, curcumin and catechin, bind directly to the receptor-binding domain of the S-protein and the angiotensin-converting enzyme 2 receptor of the host cell, by which these molecules inhibit the entry of viruses in the host cell. As a result, the animal host will survive the infection.
Collapse
Affiliation(s)
- Gajendra Kumar
- Department of Chemistry, Krishna College, Bijnor -246701, Uttar Pradesh, India
| | - Dharmendra Kumar
- Science Branch, Archeological Survey of India, Patna Zone, India
| | - Netra Pal Singh
- Department of Chemistry, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur -273009, Uttar Pradesh, India
| |
Collapse
|
12
|
Wu CY, Lin YS, Yang YH, Shu LH, Cheng YC, Liu HT. GB-2 inhibits ACE2 and TMPRSS2 expression: In vivo and in vitro studies. Biomed Pharmacother 2020; 132:110816. [PMID: 33049583 PMCID: PMC7547400 DOI: 10.1016/j.biopha.2020.110816] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/10/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022] Open
Abstract
After the first case of Coronavirus disease 2019 (COVID-19) was reported in Wuhan, COVID-19 has rapidly spread to almost all parts of world. Angiotensin converting enzyme 2 (ACE2) receptor can bind to spike protein of SARS-CoV-2. Then, the spike protein of SARS-CoV-2 can be cleaved and activated by transmembrane protease, serine 2 (TMPRSS2) of the host cells for SARS-CoV-2 infection. Therefore, ACE2 and TMPRSS2 are potential antiviral targets for treatment of prevention of SARS-CoV-2 infection. In this study, we discovered that 10-250 μg/mL of GB-2, from Tian Shang Sheng Mu of Chiayi Puzi Peitian Temple, can inhibit ACE2 mRNA expression and ACE2 and TMPRSS2 protein expression in HepG2 and 293 T cells without cytotoxicity. GB-2 treatment could decrease ACE2 and TMPRSS2 expression level of lung tissue and kidney tissue without adverse effects, including nephrotoxicity and hepatotoxicity, in animal model. In the compositions of GB-2, we discovered that 50 μg/mL of theaflavin could inhibit protein expression of ACE2 and TMPRSS2. Theaflavin could inhibit the mRNA expression of ACE2. In conclusion, our results suggest that GB-2 and theaflavin could act as potential compounds for ACE2 and TMPRSS2 inhibitors in the further clinical study.
Collapse
Affiliation(s)
- Ching-Yuan Wu
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan; School of Chinese Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| | - Yu-Shih Lin
- Department of Pharmacy, Chiayi Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan; School of Chinese Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan
| | - Li-Hsin Shu
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan
| | - Yu-Ching Cheng
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan
| | - Hung Te Liu
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Putzu, Taiwan
| |
Collapse
|
13
|
Levy E, Delvin E, Marcil V, Spahis S. Can phytotherapy with polyphenols serve as a powerful approach for the prevention and therapy tool of novel coronavirus disease 2019 (COVID-19)? Am J Physiol Endocrinol Metab 2020; 319:E689-E708. [PMID: 32755302 PMCID: PMC7518070 DOI: 10.1152/ajpendo.00298.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 02/08/2023]
Abstract
Much more serious than the previous severe acute respiratory syndrome (SARS) coronavirus (CoV) outbreaks, the novel SARS-CoV-2 infection has spread speedily, affecting 213 countries and causing ∼17,300,000 cases and ∼672,000 (∼+1,500/day) deaths globally (as of July 31, 2020). The potentially fatal coronavirus disease (COVID-19), caused by air droplets and airborne as the main transmission modes, clearly induces a spectrum of respiratory clinical manifestations, but it also affects the immune, gastrointestinal, hematological, nervous, and renal systems. The dramatic scale of disorders and complications arises from the inadequacy of current treatments and absence of a vaccine and specific anti-COVID-19 drugs to suppress viral replication, inflammation, and additional pathogenic conditions. This highlights the importance of understanding the SARS-CoV-2 mechanisms of actions and the urgent need of prospecting for new or alternative treatment options. The main objective of the present review is to discuss the challenging issue relative to the clinical utility of plants-derived polyphenols in fighting viral infections. Not only is the strong capacity of polyphenols highlighted in magnifying health benefits, but the underlying mechanisms are also stressed. Finally, emphasis is placed on the potential ability of polyphenols to combat SARS-CoV-2 infection via the regulation of its molecular targets of human cellular binding and replication, as well as through the resulting host inflammation, oxidative stress, and signaling pathways.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, Sainte-Justine University Health Center, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Edgard Delvin
- Research Centre, Sainte-Justine University Health Center, Montreal, Quebec, Canada
| | - Valérie Marcil
- Research Centre, Sainte-Justine University Health Center, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Schohraya Spahis
- Research Centre, Sainte-Justine University Health Center, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
14
|
Oliveira MB, Valentim IB, Rocha TS, Santos JC, Pires KS, Tanabe EL, Borbely KS, Borbely AU, Goulart MO. Schinus terebenthifolius Raddi extracts: From sunscreen activity toward protection of the placenta to Zika virus infection, new uses for a well-known medicinal plant. INDUSTRIAL CROPS AND PRODUCTS 2020; 152:112503. [PMID: 32346222 PMCID: PMC7186214 DOI: 10.1016/j.indcrop.2020.112503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 05/02/2023]
Abstract
Schinus terebinthifolius Raddi is a well-known medicinal plant native of South America. This species has demonstrated important biological activities such as antihypertensive and vasodilator, antimicrobial, anti-inflammatory and antioxidant. However, no studies have been, so far, reported with the fruits of S. terebinthifolius as a protector of the placenta against Zika virus infection and as sunscreen agents. The present study aimed to investigate new uses for the ethanolic fruit extracts of S. terebinthifolius, from fruits'peel (STPE) and from the whole fruits (STWFE). Zika virus (ZIKV) has been linked to several fetal malformations, such as microcephaly and other central nervous system abnormalities. Thus, the potential of these natural extracts against ZIKV infection was evaluated, using an in vitro method. The photoprotective potential, determined by spectrometry, along with phenolic content, antioxidant capacity, and chemical composition of both extracts were also evaluated. The chemical composition of the extracts was evaluated by HPLC-UV / vis. The cytotoxicity of peel and whole fruit extracts in vero E6 cell lines, in placental cell lines and placental explant cultures were evaluated by the MTT assay. The infectivity of placental cells and explants was evaluated by qRT-PCR and the effects of extracts on ZIKV infection were investigated using HTR-8/SVneo cells, pre-treated with 100 μg mL-1 of STWFE for 1 h, and infected with MR766 (AD) or PE243 (EH) ZIKV strains. STFE and STWFE were well-tolerated by both placental-derived trophoblast cell line HTR-8/SVneo as well as by term placental chorionic villi explants, which indicate absence of cytotoxicity in all analysed concentrations. Two strains of ZIKV were tested to access if pre-treatment of trophoblast cells with the STWFE would protect them against infection. Flow cytometry analysis revealed that STWFE extract greatly reduced ZIKV infection. The extracts were also photoprotective with SPF values equivalent to the standard, benzophenone-3. The formulations prepared in different concentrations of the extracts (5-10 %) had shown maximum SPF values of 32.21. STWFE represents a potential natural mixture to be used in pregnancy in order to restrain placental infection by ZIKV and might potentially protect fetus against ZIKV-related malformations. The extracts exhibited photoprotective activity and some of the phenolic compounds, mainly resveratrol, catechin and epicatechin, are active ingredients in all assayed activities. The development of biotechnological/medical products, giving extra value to products from family farming, is expected, with strong prospects for success.
Collapse
Affiliation(s)
- Monika B.S. Oliveira
- Universidade Federal de Alagoas (UFAL), Instituto de Química e Biotecnologia, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
| | - Iara B. Valentim
- Instituto Federal de Educação, Ciência e Tecnologia de Alagoas (IFAL), Rua Mizael Domingues, 75, Centro, CEP 57020-600, Maceió, AL, Brazil
| | - Tauane S. Rocha
- Universidade Federal de Alagoas (UFAL), Instituto de Química e Biotecnologia, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
| | - Jaqueline C. Santos
- Universidade Federal de Alagoas (UFAL), Instituto de Ciências Biológicas e da Saúde, Laboratório de Biologia Celular, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
| | - Keyla S.N. Pires
- Universidade Federal de Alagoas (UFAL), Instituto de Ciências Biológicas e da Saúde, Laboratório de Biologia Celular, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
| | - Eloiza L.L. Tanabe
- Universidade Federal de Alagoas (UFAL), Instituto de Ciências Biológicas e da Saúde, Laboratório de Biologia Celular, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
| | - Karen S.C. Borbely
- Universidade Federal de Alagoas (UFAL), Instituto de Ciências Biológicas e da Saúde, Laboratório de Biologia Celular, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
- Universidade Federal de Alagoas (UFAL), Faculdade de Nutrição, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
| | - Alexandre U. Borbely
- Universidade Federal de Alagoas (UFAL), Instituto de Ciências Biológicas e da Saúde, Laboratório de Biologia Celular, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
- Corresponding auhtors at: Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Campus A.C. Simões, Tabuleiro dos Martins, 57072-970, Maceió, AL, Brazil.
| | - Marília O.F. Goulart
- Universidade Federal de Alagoas (UFAL), Instituto de Química e Biotecnologia, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, 57072-970, Maceió, AL, Brazil
- Corresponding auhtors at: Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Campus A.C. Simões, Tabuleiro dos Martins, 57072-970, Maceió, AL, Brazil.
| |
Collapse
|
15
|
Yadav P, Choudhury S, Barua S, Khandelwal N, Kumar N, Shukla A, Garg SK. Polyalthia longifolia leaves methanolic extract targets entry and budding of viruses-an in vitro experimental study against paramyxoviruses. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112279. [PMID: 31600562 DOI: 10.1016/j.jep.2019.112279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 06/04/2019] [Accepted: 10/04/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Synthetic antiviral drugs have several limitations including high cost. Thus research on antiviral property of medicinal plants is continuously gaining importance. Polyalthia longifolia possesses several medicinal properties and has been used in traditional ayurvedic medicine for treatment of dermatological ailments as kushta, visarpa/herpes virus infection and also to treat pyrexia of unknown origin as mentioned in Visarpa Chikitsa. AIM OF THE STUDY Keeping in view the cytotoxic, anti-cancer activity and antiviral efficacy of Polyalthia longifolia against herpes, present study was undertaken to evaluate the in vitro antiviral activity of methanolic extract of Polyalthia longifolia leaves, if any, and to unravel the possible target(s)/mechanism of action. MATERIAL AND METHODS Antiviral activity of Polyalthia longifolia methanolic extract was studied using Vero cell lines against paramyxoviruses, namely-peste des petits ruminants virus (PPRV) and Newcastle disease virus (NDV). Cytotoxicity of the test extract was evaluated employing MTT assay. Virucidal activity, and viral-attachment, virus entry and release assays were determined in Vero cells using standard experimental protocols. The viral RNA in the virus-infected cells was quantified by qRT-PCR. RESULTS At non-cytotoxic concentration, methanolic extract of Polyalthia longifolia leaves was found to inhibit the replication of PPRV and NDV at viral entry and budding level, whereas other steps of viral life cycle such as attachment and RNA synthesis remained unaffected. CONCLUSIONS Polyalthia longifolia leaves extract possesses promising antiviral activity against paramyxoviruses and acts by inhibiting the entry and budding of viruses; and this plant extract evidently possesses excellent and promising potential for development of effective herbal antiviral drug.
Collapse
Affiliation(s)
- Prashant Yadav
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, India.
| | - Soumen Choudhury
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, India.
| | - Sanjay Barua
- National Centre for Veterinary Type Culture Collections (NCVTCC), ICAR-National Research Centre on Equines, Hisar, Haryana, India.
| | - Nitin Khandelwal
- National Centre for Veterinary Type Culture Collections (NCVTCC), ICAR-National Research Centre on Equines, Hisar, Haryana, India.
| | - Naveen Kumar
- National Centre for Veterinary Type Culture Collections (NCVTCC), ICAR-National Research Centre on Equines, Hisar, Haryana, India; Division of Goat Health, ICAR-Central Institute for Research on Goats, Makhdoom, Mathura, India.
| | - Amit Shukla
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, India.
| | - Satish K Garg
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, India.
| |
Collapse
|
16
|
Multilevel structure-activity profiling reveals multiple green tea compound families that each modulate ubiquitin-activating enzyme and ubiquitination by a distinct mechanism. Sci Rep 2019; 9:12801. [PMID: 31488855 PMCID: PMC6728334 DOI: 10.1038/s41598-019-48888-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022] Open
Abstract
We developed and implemented a reconstituted system to screen for modulators of the ubiquitination of proliferating cell nuclear antigen, a process that activates pathways of DNA damage tolerance and drug resistance. We identified the primary putatively health-beneficial green tea polyphenol epigallocatechin gallate (EGCG) and certain related small molecules as potent inhibitors of ubiquitination. EGCG directly and reversibly targets the ubiquitin-activating enzyme Uba1, blocking formation of the Uba1~ubiquitin thioester conjugate and thus ubiquitination and in the cell. Structure–activity relationship profiles across multiple biochemical and cellular assays for a battery of EGCG analogues revealed distinct chemical and mechanism-of-action clusters of molecules, with catechin gallates, alkyl gallates, and myricetin potently inhibiting ubiquitination. This study defines a number of related though distinct first-in-class inhibitors of ubiquitination, each series with its own unique activity pattern and mechanistic signature.
Collapse
|
17
|
Hao X, Zhang W, Zhao F, Liu Y, Qian W, Wang Y, Wang L, Zeng J, Yang Y, Wang X. Discovery of Plant Viruses From Tea Plant ( Camellia sinensis (L.) O. Kuntze) by Metagenomic Sequencing. Front Microbiol 2018; 9:2175. [PMID: 30254625 PMCID: PMC6141721 DOI: 10.3389/fmicb.2018.02175] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022] Open
Abstract
The tea plant (Camellia sinensis (L.) O. Kuntze) is an economically important woody species. In this study, we collected 26 tea plant samples with typical discoloration symptoms from different tea gardens and performed metagenomic analysis based on next-generation sequencing. Homology annotation and PCR sequencing validation finally identified seven kinds of plant viruses from tea plant. Based on abundance distribution analysis, the two most abundant plant viruses were highlighted. Genetic characterization suggested that they are two novel virus species with relatively high homology to Blueberry necrotic ring blotch virus and American plum line pattern virus. We named the newly discovered viruses tea plant necrotic ring blotch virus (TPNRBV) and tea plant line pattern virus (TPLPV). Evolutionary relationship analysis indicated that TPNRBV and TPLPV should be grouped into the Blunervirus and the Ilarvirus genera, respectively. TPLPV might have same genome activation process with known ilarviruses based on sequence analysis. Moreover, specific primers for both viruses detection were designed and validated. The symptoms and ultrastructure of TPNRBV infected leaves were first recorded. Virus detections in the symptomatic and asymptomatic tissues from field plants showing tea plant necrotic ring blotch disease suggest that TPNRBV has a systemic movement feature. In summary, we first identified seven kinds of putative plant viruses by metagenomic analysis and report two novel viruses being latent pathogens to tea plant. The results will advance our understanding of tea plant virology and have significance for the genetic breeding of tea plants in the future.
Collapse
Affiliation(s)
- Xinyuan Hao
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Weifu Zhang
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Fumei Zhao
- Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Ying Liu
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Wenjun Qian
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Yuchun Wang
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Lu Wang
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Jianming Zeng
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Yajun Yang
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| | - Xinchao Wang
- National Center for Tea Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, China
| |
Collapse
|
18
|
Nakano S, Megro SI, Hase T, Suzuki T, Isemura M, Nakamura Y, Ito S. Computational Molecular Docking and X-ray Crystallographic Studies of Catechins in New Drug Design Strategies. Molecules 2018; 23:E2020. [PMID: 30104534 PMCID: PMC6222539 DOI: 10.3390/molecules23082020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/09/2018] [Accepted: 08/11/2018] [Indexed: 12/16/2022] Open
Abstract
Epidemiological and laboratory studies have shown that green tea and green tea catechins exert beneficial effects on a variety of diseases, including cancer, metabolic syndrome, infectious diseases, and neurodegenerative diseases. In most cases, (-)-epigallocatechin gallate (EGCG) has been shown to play a central role in these effects by green tea. Catechins from other plant sources have also shown health benefits. Many studies have revealed that the binding of EGCG and other catechins to proteins is involved in its action mechanism. Computational docking analysis (CMDA) and X-ray crystallographic analysis (XCA) have provided detailed information on catechin-protein interactions. Several of these studies have revealed that the galloyl moiety anchors it to the cleft of proteins through interactions with its hydroxyl groups, explaining the higher activity of galloylated catechins such as EGCG and epicatechin gallate than non-galloylated catechins. In this paper, we review the results of CMDA and XCA of EGCG and other plant catechins to understand catechin-protein interactions with the expectation of developing new drugs with health-promoting properties.
Collapse
Affiliation(s)
- Shogo Nakano
- School of Food and Nutritional Sciences, Shizuoka University, Yada, Shizuoka 422-8526, Japan.
| | - Shin-Ichi Megro
- Biological Science Research, Kao Corporation, Ichikai-machi, Haga-gun, Tochigi 321-3497, Japan.
| | - Tadashi Hase
- Research and Development, Core Technology, Kao Corporation, Sumida, Tokyo 131-8501, Japan.
| | - Takuji Suzuki
- Faculty of Education, Art and Science, Yamagata University, Yamagata 990-8560, Japan.
| | - Mamoru Isemura
- School of Food and Nutritional Sciences, Shizuoka University, Yada, Shizuoka 422-8526, Japan.
| | - Yoriyuki Nakamura
- School of Food and Nutritional Sciences, Shizuoka University, Yada, Shizuoka 422-8526, Japan.
| | - Sohei Ito
- School of Food and Nutritional Sciences, Shizuoka University, Yada, Shizuoka 422-8526, Japan.
| |
Collapse
|