1
|
Reilly CB, Moore J, Lightbown S, Paul A, Bernier SG, Carlson KE, Ingber DE. Broad-spectrum coronavirus inhibitors discovered by modeling viral fusion dynamics. Front Mol Biosci 2025; 12:1575747. [PMID: 40443526 PMCID: PMC12119275 DOI: 10.3389/fmolb.2025.1575747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025] Open
Abstract
Development of oral, broad-spectrum therapeutics targeting SARS-CoV-2, its variants, and related coronaviruses could curb the spread of COVID-19 and avert future pandemics. We created a novel computational discovery pipeline that employed molecular dynamics simulation (MDS), artificial intelligence (AI)-based docking predictions, and medicinal chemistry to design viral entry inhibitors that target a conserved region in the SARS-CoV-2 spike (S) protein that mediates membrane fusion. DrugBank library screening identified the orally available, FDA-approved AXL kinase inhibitor bemcentinib as binding this site and we demonstrated that it inhibits viral entry in a kinase-independent manner. Novel analogs predicted to bind to the same region and disrupt S protein conformational changes were designed using MDS and medicinal chemistry. These compounds significantly suppressed SARS-CoV-2 infection and blocked the entry of S protein-bearing pseudotyped α,β,γ,δ,ο variants as well as SARS CoV and MERS-CoV in human ACE2-expressing or DPP4-expressing cells more effectively than bemcentinib. When administered orally, the optimized lead compound also significantly inhibited SARS-CoV2 infection in mice. This computational design strategy may accelerate drug discovery for a broad range of applications.
Collapse
Affiliation(s)
- Charles B. Reilly
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| | - Joel Moore
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| | - Shanda Lightbown
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| | - Austin Paul
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| | - Sylvie G. Bernier
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| | - Kenneth E. Carlson
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
- Vascular Biology Program and Department of Surgery, Harvard Medical School and Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
2
|
Barzin Tond S, Abolghasemi S, Khatami SH, Ehtiati S, Zarei T, Shateri S, Mahmoodi Baram S, Yarahmadi S, Fallah S, Salmani F, Shahmohammadi MR, Khajavirad N, Tafakhori A, Riazi A, Karima S. Boswellic Acids Reduce Systemic Inflammation in Patients with Moderate COVID-19 Through Modulation of NF-κB Pathway. J Diet Suppl 2025; 22:365-381. [PMID: 40012185 DOI: 10.1080/19390211.2025.2468484] [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] [Indexed: 02/28/2025]
Abstract
Prevention and/or management of the dysregulated immune response in patients with COVID-19 is expected to help in the treatment of COVID-19. Boswellic acids (BAs) have great therapeutic potential because they have anti-inflammatory and immunomodulatory effects. Here, we aimed to investigate the mechanism of action of a BA formulation, Inflawell syrup, which was previously shown to be effective in reducing disease symptoms in patients who suffer from mild to moderate COVID-19. Patients with mild to moderate COVID-19 were treated with either Inflawell containing boswellic acids or a placebo for 14 days. The serum levels of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-8, IL-1α, IL-17, IL-1Ra, and Monocyte Chemoattractant Protein-1 (MCP-1), were measured both at study onset and on day 14 after treatment started. In addition, to further investigate the signaling pathway(s) underlying the changes in cytokine levels, we evaluated the expression of tumor necrosis factor receptor 1 (TNFR1), tumor necrosis factor receptor 2 (TNFR2), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 mRNAs and phospho-inhibitor of nuclear factor kappa B (IκB) and IκB proteins. In our study, a significant decrease in the serum levels of IL-1α (p < .009), IL-8 (p < .04), TNF-α (p < .0001), and MCP-1 (p < .007) was detected in patients treated with Inflawell. Additionally, our data revealed a decrease in phospho-IκB protein levels (p < .02) and NF-κB p65 mRNA levels (p < .002), whereas the amount of IκB protein (p < .01) in the Inflawell group was significantly greater than that in the placebo group. Furthermore, despite the decreasing trend in the expression of TNFR1 and TNFR2 in the Inflawell group, there was no statistically significant difference compared with that in the placebo group. In general, treatment with Inflawell syrup led to a lower level of proinflammatory cytokines and a decrease in the activity of the TNF-α/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Sepideh Barzin Tond
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Sara Abolghasemi
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medial Sciences (SBMU), Tehran, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayebe Zarei
- Clinical Trial Department, Behbalin Co., Ltd, Tehran, Iran
| | - Somayyeh Shateri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | | | - Sahar Yarahmadi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soudabeh Fallah
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Salmani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Mohammad Reza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Nasim Khajavirad
- Internal Medicine Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| |
Collapse
|
3
|
Shu C, Jiao W, Cui K, Cao J, Jiang W. Ursolic Acid Induces Multifaceted Defense Responses Against Postharvest Blue Mold Rot in Apple Fruit. Foods 2025; 14:761. [PMID: 40077464 PMCID: PMC11899365 DOI: 10.3390/foods14050761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The disease resistance and defense mechanisms induced by ursolic acid (UA) in apple fruit were studied in this paper. UA was directly mixed with potato dextrose agar and broth media to assay its antifungal activity in vitro. The results showed that UA exerted inherent antifungal activity and directly inhibited the in vitro growth and spore germination of Penicillium expansum. Its half-maximal inhibitory concentration for hyphal growth was 175.6 mg L-1. Apple fruit were immersed in UA solution, followed by inoculation with P. expansum, to measure their disease response. The results demonstrated that UA induced significant disease resistance in apple fruit and that its mechanisms are multifaceted and associated with defensive and antioxidative enzymes and the phenylpropanoid pathway. Chitinase, β-1,3-glucanase, peroxidase, and polyphenol oxidase were activated and maintained at relatively high levels. The activities of enzymes and their metabolites in the phenylpropanoid pathway, including phenylalanine ammonia-lyase, cinnamate-4-hydroxylase, and 4-coumarate coenzyme A ligase were significantly increased; accordingly, total phenolics, flavonoid, and lignin contents were significantly increased. The activities of superoxide dismutase, ascorbate peroxidase, and glutathione reductase were enhanced upon UA treatment, while catalase activity was suppressed, which regulates hydrogen peroxide accumulation to defend against pathogens. These results suggest that UA induces defense responses against postharvest blue mold rot in apple fruit and that it may be a promising elicitor to induce fruit disease resistance to control postharvest decay.
Collapse
Affiliation(s)
- Chang Shu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (C.S.); (J.C.)
| | - Wenxiao Jiao
- College of Food Science and Engineering, Qilu University of Technology, Jinan 250353, China;
| | - Kuanbo Cui
- Agricultural Mechanization Institute, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China;
| | - Jiankang Cao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (C.S.); (J.C.)
| | - Weibo Jiang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (C.S.); (J.C.)
| |
Collapse
|
4
|
Kgosiemang IKR, Lefojane R, Adegoke AM, Ogunyemi O, Mashele SS, Sekhoacha MP. Pharmacological Significance, Medicinal Use, and Toxicity of Extracted and Isolated Compounds from Euphorbia Species Found in Southern Africa: A Review. PLANTS (BASEL, SWITZERLAND) 2025; 14:469. [PMID: 39943031 PMCID: PMC11821031 DOI: 10.3390/plants14030469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/30/2024] [Accepted: 01/19/2025] [Indexed: 02/16/2025]
Abstract
This study documents the Euphorbiaceae family of plants in Southern Africa, with a focus on their traditional medicinal applications, pharmacological properties, toxicity, and active secondary metabolites. A review of the literature from scientific journals, books, dissertations, and conference papers spanning from 1962 to 2023 was conducted for 15 Euphorbia species. Recent findings indicate that specific compounds found in Euphorbia plants exhibit significant biological and pharmacological properties. However, the white sticky latex sap they contain is highly toxic, although it may also have medicinal applications. Phytochemical analyses have demonstrated that these plants exhibit beneficial effects, including antibacterial, antioxidant, antiproliferative, anticancer, anti-inflammatory, antiviral, antifungal, and anti-HIV activities. Key phytochemicals such as euphol, cycloartenol, tirucallol, and triterpenoids contribute to their therapeutic efficacy, along with various proteins like lectin and lysozyme. Despite some Euphorbiaceae species undergoing screening for medicinal compounds, many remain insufficiently examined, highlighting a critical gap in the research literature. Given their historical usage, further investigations are essential to evaluate the medicinal significance of Euphorbia species through detailed studies of isolated compounds and their pharmacokinetics and pharmacodynamics. This research will serve as a valuable resource for future inquiries into the benefits of lesser-studied Euphorbia species.
Collapse
Affiliation(s)
- Ipeleng Kopano Rosinah Kgosiemang
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, South Africa; (I.K.R.K.); (R.L.); (S.S.M.)
| | - Relebohile Lefojane
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, South Africa; (I.K.R.K.); (R.L.); (S.S.M.)
| | - Ayodeji Mathias Adegoke
- Department of Pharmacology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa;
- Cancer Research and Molecular Biology Laboratories, College of Medicine, University of Ibadan, Ibadan 200005, Nigeria
| | - Oludare Ogunyemi
- Nutritional and Industrial Biochemistry Research Unit, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan 200005, Nigeria;
| | - Samson Sitheni Mashele
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, South Africa; (I.K.R.K.); (R.L.); (S.S.M.)
| | - Mamello Patience Sekhoacha
- Department of Pharmacology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa;
| |
Collapse
|
5
|
Duan Y, Pei K, Liu X, Zhang X, Song P, Tu S, Zhu H, Cai H. A comprehensive pharmacokinetic strategy for systematic evaluation of whole interaction of different constituents in Astragali Radix -Fructus Corni to improve diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119159. [PMID: 39603397 DOI: 10.1016/j.jep.2024.119159] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/08/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragali-Radix (the dried root of Astragalus mongholicus Bunge, AR) - Fructus Corni (the dried ripe fruit of Cornus officinalis Sieb. et Zucc., FC) has been used as a herb-pair remedy to treat diabetic kidney disease (DKD) for hundred years. Polysaccharides, saponins, and flavonoids in AR, and the iridoid glycosides in FC were deemed as the main bioactive constituents that can offer beneficial nephroprotective activities. A systematic evaluation of the nephroprotective effects of AR-FC herb pair, the main bioactive constituents extracted from the herb pair, and their combinations in different ratios was performed, CG6 (polysaccharides, flavonoids, saponins, and iridoid glycosides, in a ratio of 2:3:1:2) as the best compatibility proportion was screened out in our previous study. AIM OF THE STUDY This study aimed to investigate the pharmacokinetic characteristics of AR-FC herb-pair in DKD rats, and explore the interactions between constituents from AR-FC and the rational compatibility of different constituents. MATERIALS AND METHODS The protective effect of AR-FC and CG6 on renal injury caused by DKD was first verified by histopathological examination. Then, an analytical method based on UHPLC-Q-TOF-MS and UHPLC-QqQ-MS/MS for qualitative and quantitative metabolites without reference standards was established and applied to pharmacokinetic (PK) studies in following different aspects: between single groups (polysaccharides, flavonoids, saponins and iridoid glycosides) and compatibility groups (AR-FC, CG6), in normal and DKD rats, in single-dose administration and long-term administration. RESULTS Pathological observations confirmed that AR-FC could improve renal injury in DKD rats. PK profiles of nine prototypes and four metabolites in various groups were obtained, revealing the compatibility of multiple constitutes, pathological states, and long-term administration could alter PK characteristics of the main components from AR-FC, and promoting the absorption of them (Cmax, AUC0-t, and AUC0-t increased). Notably, co-administration of iridoid glycosides could significantly increase the absorption of flavonoids and saponins in vivo. The pharmacokinetics based on homologous compounds revealed that saponins first acted, then its initial metabolites affected flavonoids, and ultimately the metabolites of flavonoids influenced iridoid glycosides. CONCLUSION This study demonstrated the existence of interactions between constituents from AR-FC herb-pair and the importance of their rational compatibility. It provides experimental evidence for developing a therapeutic agent based on AR-FC (especially CG6) to treat DKD. It is also expected to provide a reference for the multi-component pharmacokinetic study of other herbal medicines.
Collapse
Affiliation(s)
- Yu Duan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Ke Pei
- School of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, PR China.
| | - Xue Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Xia Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Peixiang Song
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Sicong Tu
- Brain & Mind Centre, Faculty of Medicine & Health, The University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Hui Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Hao Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
6
|
Li Y, Zhu X, Xiong W, Zhao Q, Zhou Y, Guo Y, Liu B, Li M, Chen Q, Jiang X, Qi Y, Ye Q, Deng G. Brain-targeted ursolic acid nanoparticles for anti-ferroptosis therapy in subarachnoid hemorrhage. J Nanobiotechnology 2024; 22:641. [PMID: 39425081 PMCID: PMC11490124 DOI: 10.1186/s12951-024-02866-x] [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: 03/27/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a life -threatening cerebrovascular disease, where early brain injury (EBI) stands as a primary contributor to mortality and unfavorable patient outcomes. Neuronal ferroptosis emerges as a key pathological mechanism underlying EBI in SAH. Targeting ferroptosis for therapeutic intervention in SAH holds significant promise as a treatment strategy. METHODS SAH model was induced via intravascular puncture and quantitatively assessed the presence of neuronal ferroptosis in the early phase of SAH using FJC staining, Prussian blue staining, as well as malondialdehyde (MDA) and glutathione (GSH) measurements. Hyaluronic acid-coated ursolic acid nanoparticles (HA-PEG-UA NPs) were prepared using the solvent evaporation method. We investigated the in vivo distribution of HA-PEG-UA NPs in SAH model through IVIS and fluorescence observation, and examined their impact on short-term neurological function and cortical neurological injury. Finally, we assessed the effect of UA on the Nrf-2/SLC7A11/GPX4 axis via Western Blot analysis. RESULTS We successfully developed self-assembled UA NPs with hyaluronic acid to target the increased CD44 expression in the SAH-afflicted brain. The resulting HA-PEG-UA NPs facilitated delivery and enrichment of UA within the SAH-affected region. The targeted delivery of UA to the SAH region can effectively inhibit neuronal ferroptosis, improve neurological deficits, and prognosis in mice. Its mechanism of action is associated with the activation of the Nrf-2/SLC7A11/GPX4 signaling pathway. CONCLUSIONS Brain-targeted HA-PEG-UA NPs was successfully developed and hold the potential to enhance SAH prognosis by limiting neuronal ferroptosis via modulation of the Nrf-2/SLC7A11/GPX4 signal.
Collapse
Affiliation(s)
- Yong Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Xiong
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingyu Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Youdong Zhou
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yujia Guo
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yangzhi Qi
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China.
- Sydney Dental School, The University of Sydney, Camperdown, NSW, 2050, Sydney, Australia.
| | - Gang Deng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
7
|
Shekunov EV, Efimova SS, Kever LV, Ishmanov TF, Ostroumova OS. Lipid Selectivity of Membrane Action of the Fragments of Fusion Peptides of Marburg and Ebola Viruses. Int J Mol Sci 2024; 25:9901. [PMID: 39337389 PMCID: PMC11432738 DOI: 10.3390/ijms25189901] [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: 07/26/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The life cycle of Ebola and Marburg viruses includes a step of the virion envelope fusion with the cell membrane. Here, we analyzed whether the fusion of liposome membranes under the action of fragments of fusion peptides of Ebola and Marburg viruses depends on the composition of lipid vesicles. A fluorescence assay and electron microscopy were used to quantify the fusogenic activity of the virus fusion peptides and to identify the lipid determinants affecting membrane merging. Differential scanning calorimetry of lipid phase transitions revealed alterations in the physical properties of the lipid matrix produced by virus fusion peptides. Additionally, we found that plant polyphenols, quercetin, and myricetin inhibited vesicle fusion induced by the Marburg virus fusion peptide.
Collapse
Affiliation(s)
- Egor V Shekunov
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Svetlana S Efimova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Lyudmila V Kever
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Tagir F Ishmanov
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Olga S Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| |
Collapse
|
8
|
Sari IP, Ortiz CLD, Yang LW, Chen MH, Perng MD, Wu TY. Development of Fusion-Based Assay as a Drug Screening Platform for Nipah Virus Utilizing Baculovirus Expression Vector System. Int J Mol Sci 2024; 25:9102. [PMID: 39201788 PMCID: PMC11354753 DOI: 10.3390/ijms25169102] [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: 06/06/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Nipah virus (NiV) is known to be a highly pathogenic zoonotic virus, which is included in the World Health Organization Research & Development Blueprint list of priority diseases with up to 70% mortality rate. Due to its high pathogenicity and outbreak potency, a therapeutic countermeasure against NiV is urgently needed. As NiV needs to be handled within a Biological Safety Level (BSL) 4 facility, we had developed a safe drug screening platform utilizing a baculovirus expression vector system (BEVS) based on a NiV-induced syncytium formation that could be handled within a BSL-1 facility. To reconstruct the NiV-induced syncytium formation in BEVS, two baculoviruses were generated to express recombinant proteins that are responsible for inducing the syncytium formation, including one baculovirus exhibiting co-expressed NiV fusion protein (NiV-F) and NiV attachment glycoprotein (NiV-G) and another exhibiting human EphrinB2 protein. Interestingly, syncytium formation was observed in infected insect cells when the medium was modified to have a lower pH level and supplemented with cholesterol. Fusion inhibitory properties of several compounds, such as phytochemicals and a polysulfonated naphthylamine compound, were evaluated using this platform. Among these compounds, suramin showed the highest fusion inhibitory activity against NiV-induced syncytium in the baculovirus expression system. Moreover, our in silico results provide a molecular-level glimpse of suramin's interaction with NiV-G's central hole and EphrinB2's G-H loop, which could be the possible reason for its fusion inhibitory activity.
Collapse
Affiliation(s)
- Indah Permata Sari
- Institute of Molecular Medicine, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (I.P.S.); (M.-H.C.)
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| | - Christopher Llynard D. Ortiz
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Taipei 11529, Taiwan; (C.L.D.O.); (L.-W.Y.)
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Lee-Wei Yang
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Taipei 11529, Taiwan; (C.L.D.O.); (L.-W.Y.)
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ming-Hsiang Chen
- Institute of Molecular Medicine, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (I.P.S.); (M.-H.C.)
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (I.P.S.); (M.-H.C.)
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| |
Collapse
|
9
|
Kim H, Kang Y, Kim S, Park D, Heo SY, Yoo JS, Choi I, N MPA, Ahn JW, Yang JS, Bak N, Kim KK, Lee JY, Choi YK. The host protease KLK5 primes and activates spike proteins to promote human betacoronavirus replication and lung inflammation. Sci Signal 2024; 17:eadn3785. [PMID: 39163389 DOI: 10.1126/scisignal.adn3785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/03/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024]
Abstract
Coronaviruses rely on host proteases to activate the viral spike protein, which facilitates fusion with the host cell membrane and the release of viral genomic RNAs into the host cell cytoplasm. The distribution of specific host proteases in the host determines the host, tissue, and cellular tropism of these viruses. Here, we identified the kallikrein (KLK) family member KLK5 as a major host protease secreted by human airway cells and exploited by multiple human betacoronaviruses. KLK5 cleaved both the priming (S1/S2) and activation (S2') sites of spike proteins from various human betacoronaviruses in vitro. In contrast, KLK12 and KLK13 displayed preferences for either the S2' or S1/S2 site, respectively. Whereas KLK12 and KLK13 worked in concert to activate SARS-CoV-2 and MERS-CoV spike proteins, KLK5 by itself efficiently activated spike proteins from several human betacoronaviruses, including SARS-CoV-2. Infection of differentiated human bronchial epithelial cells (HBECs) with human betacoronaviruses induced an increase in KLK5 that promoted virus replication. Furthermore, ursolic acid and other related plant-derived triterpenoids that inhibit KLK5 effectively suppressed the replication of SARS-CoV, MERS-CoV, and SARS-CoV-2 in HBECs and mitigated lung inflammation in mice infected with MERS-CoV or SARS-CoV-2. We propose that KLK5 is a pancoronavirus host factor and a promising therapeutic target for current and future coronavirus-induced diseases.
Collapse
Affiliation(s)
- Hyunjoon Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Yeonglim Kang
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Semi Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Dongbin Park
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Seo-Young Heo
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Ji-Seung Yoo
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Isaac Choi
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Monford Paul Abishek N
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Jae-Woo Ahn
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Jeong-Sun Yang
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health (KNIH), 187 Osongsaengmyeong2-ro, Heungdeok-gu, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Nayeon Bak
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
- Department of Metabiohealth, Sungkyun Convergence Institute, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Kyeong Kyu Kim
- Department of Metabiohealth, Sungkyun Convergence Institute, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health (KNIH), 187 Osongsaengmyeong2-ro, Heungdeok-gu, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Young Ki Choi
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea
- Department of Metabiohealth, Sungkyun Convergence Institute, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
10
|
Shen P, Jiang X, Kuang Y, Wang W, Raj R, Wang W, Zhu Y, Zhang X, Yu B, Zhang J. Natural triterpenoid-aided identification of the druggable interface of HMGB1 occupied by TLR4. RSC Chem Biol 2024; 5:751-762. [PMID: 39092445 PMCID: PMC11289874 DOI: 10.1039/d4cb00062e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/04/2024] [Indexed: 08/04/2024] Open
Abstract
HMGB1 interacts with TLR4 to activate the inflammatory cascade response, contributing to the pathogenesis of endogenous tissue damage and infection. The immense importance of HMGB1-TLR4 interaction in the immune system has made its binding interface an area of significant interest. To map the binding interface of HMGB1 occupied by TLR4, triterpenoids that disrupt the HMGB1-TLR4 interaction and interfere with HMGB1-induced inflammation were developed. Using the unique triterpenoid PT-22 as a probe along with photoaffinity labeling and site-directed mutagenesis, we found that the binding interface of HMGB1 was responsible for the recognition of TLR4 located on the "L" shaped B-box with K114 as a crucial hot-spot residue. Amazingly, this highly conserved interaction surface overlapped with the antigen-recognition epitope of an anti-HMGB1 antibody. Our findings propose a novel strategy for better understanding the druggable interface of HMGB1 that interacts with TLR4 and provide insights for the rational design of HMGB1-TLR4 PPI inhibitors to fine tune immune responses.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Yi Kuang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Weiwei Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine Nanjing 210046 P. R. China
| | - Richa Raj
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
| | - Wei Wang
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago Chicago IL USA
| | - Yuyuan Zhu
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 P. R. China
| | - Xiaochun Zhang
- School of Pharmaceutical Sciences, Tsinghua University Beijing 100084 P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University Nanjing 211198 P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 P. R. China +86-25-86185158 +86-25-86185157
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University Nanjing 211198 P. R. China
| |
Collapse
|
11
|
Zhong M, Huang QJ, Bao YB, Wang JN, Mi X, Chang H, Yang Y. An oleanic acid decorated gold nanorod for highly efficient inhibition of hemagglutinin and visible rapid detection of the influenza virus. Eur J Med Chem 2024; 272:116469. [PMID: 38704939 DOI: 10.1016/j.ejmech.2024.116469] [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: 03/21/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Accurate diagnosis and effective antiviral treatments are urgently needed for the prevention and control of flu caused by influenza viruses. In this study, a novel oleanic acid (OA) functionalized gold nanorod OA-AuNP was prepared through a convenient ligand-exchange reaction. As hemagglutinin (HA) on the viral surface binds strongly to the multiple OA molecules on the surface of the nanoparticle, the prepared OA-AuNP was found to exhibit potent antiviral activity against a wide range of influenza A virus strains. Furthermore, the change in color resulting from the specific binding between HA and OA and the resultant aggregation of the OA-AuNP can be visually observed or measured by UV-vis spectra with a detection limit of 2 and 0.18 hemagglutination units (HAU), respectively, which is comparable to the commercially available influenza colloid gold rapid diagnostic kits. These findings demonstrate the potential of the OA-AuNP for the development of novel multivalent antiviral conjugates and the diagnosis of influenza virus.
Collapse
Affiliation(s)
- Ming Zhong
- Shaoguan University, Shaoguan, Guangdong Province, 512005, China
| | - Qian-Jiong Huang
- Shaoguan University, Shaoguan, Guangdong Province, 512005, China
| | - Yan-Bin Bao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Jia-Ning Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Xue Mi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Hao Chang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China.
| | - Yang Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China.
| |
Collapse
|
12
|
Shu C, Sun X, Cao J, Droby S, Jiang W. Antifungal efficiency and mechanisms of ethyl ferulate against postharvest pathogens. Int J Food Microbiol 2024; 417:110710. [PMID: 38643598 DOI: 10.1016/j.ijfoodmicro.2024.110710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
Postharvest loss caused by a range of pathogens necessitates exploring novel antifungal compounds that are safe and efficient in managing the pathogens. This study evaluated the antifungal activity of ethyl ferulate (EF) and explored its mechanisms of action against Alternaria alternata, Aspergillus niger, Botrytis cinerea, Penicillium expansum, Penicillium digitatum, Geotrichum candidum and evaluated its potential to inhibit postharvest decay. The results demonstrated that EF exerts potent antifungal activity against a wide board of postharvest pathogens. Results also revealed that its antifungal mechanism is multifaceted: EF may be involved in binding to and disturbing the integrity of the fungal plasma membrane, causing leakage of intracellular content and losing normal morphology and ultrastructure. EF also induced oxidative stress in the pathogen, causing membrane lipid peroxidation and malondialdehyde accumulation. EF inhibited the critical gene expression of the pathogen, affecting its metabolic regulation, antioxidant metabolism, and cell wall degrading enzymes. EF exhibited antifungal inhibitory activity when applied directly into peel wounds or after incorporation with chitosan coating. Due to its wide board and efficient antifungal activity, EF has the potential to provide a promising alternative to manage postharvest decay.
Collapse
Affiliation(s)
- Chang Shu
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, China; United States Department of Agriculture, Agricultural Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, 64 Nowelo Street, Hilo, HI 96720, USA; Oak Ridge Institute for Science and Education, 1299 Bethel Valley Road, Oak Ridge, TN 37830, USA
| | - Xiuxiu Sun
- United States Department of Agriculture, Agricultural Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, 64 Nowelo Street, Hilo, HI 96720, USA
| | - Jiankang Cao
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, China
| | - Samir Droby
- Department of Postharvest Science, Agricultural Research Organization, the Volcani Center, 68 Ha Maccabim Road, Rishon LeZion 7505101, Israel
| | - Weibo Jiang
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, China.
| |
Collapse
|
13
|
Ali MR, Mahmud S, Faruque MO, Hossain MI, Hossain MA, Kibria KK. Investigation of the vaccine potential of an in silico designed FepA peptide vaccine against Shigella flexneri in mice model. Vaccine X 2024; 18:100493. [PMID: 38812954 PMCID: PMC11134883 DOI: 10.1016/j.jvacx.2024.100493] [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: 01/20/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Background Shigellosis is one of the significant causes of diarrhea in Bangladesh. It is a global health problem; approximately 1.3 million people die yearly from Shigellosis. The current treatment method, using different antibiotics against Shigellosis is ineffective. Moreover, it becomes a worrying situation due to the emergence of antibiotic-resistant pathogenic microbes responsible for these diarrheal diseases. Methodology Previous immunoinformatics study predicted a potential peptide from the Ferric enterobactin protein (FepA) of Shigella spp. In this study, we have chemically synthesized the FepA peptide. As a highly immunogenic, FepA peptide conjugated with KLH has been tested in mice model with complete and incomplete adjuvants as a vaccine candidate. Results Immunological analysis showed that all vaccinated mice were immunologically boosted, which was statistically significant (P-value 0.0325) compared to control mice. Immunological analysis for bacterial neutralization test result was also statistically significant (P-value 0.0468), where each ELISA plate was coated with 1 × 107S. flexneri cells. The Challenge test with 1 × 1012S. flexneri cells to each vaccinated and controlled mice showed that 37.5 % of control (non-vaccinated) mice died within seven days after the challenge was given while 100 % of vaccinated mice remained strong and stout. The analyses of the post-challenge weight loss of the mice were also significant (P-value 0.0367) as the weight loss percentage in control mice was much higher than in the vaccinated mice. The pathological and phenotypic appearances of vaccinated mice were also clearly differentiable compared with control mice. Thus all these immunological analysis and pathological appearances directly supported our FepA peptide as a potential immune booster. Conclusion This study provides evidence that the FepA peptide is a highly immunogenic vaccine candidate against S. flexneri. Therefore, these findings inspire future trials for the evaluation of the suitability of this vaccine candidate against Shigellosis.
Collapse
Affiliation(s)
- Md. Rayhan Ali
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail-1902, Bangladesh
| | - Shahin Mahmud
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail-1902, Bangladesh
| | - Md. Omar Faruque
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail-1902, Bangladesh
| | - Md. Imam Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail-1902, Bangladesh
| | - Mohammed Akhter Hossain
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria 3052, Australia
| | - K.M. Kaderi Kibria
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail-1902, Bangladesh
| |
Collapse
|
14
|
Souza SCR, Pinheiro RR, Peixoto RM, de Sousa ALM, Andrioli A, Lima AMC, Mendes BKM, Magalhães NMDA, Amaral GP, Teixeira MFDS. In vivo evaluation of the antiretroviral activity of Melia azedarach against small ruminant lentiviruses in goat colostrum and milk. Braz J Microbiol 2024; 55:875-887. [PMID: 38010582 PMCID: PMC10920544 DOI: 10.1007/s42770-023-01174-0] [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: 05/30/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
This study aimed to evaluate in vivo the use of the extract from the leaves of Melia azedarach in the ethyl acetate fraction at a concentration of 150 µg/mL as an antiretroviral treatment against small ruminant lentiviruses (SRLV) in goat colostrum, and milk with a 90-min action. Two groups of six kids were treated with the extract. One group received three supplies of colostrum from does naturally positive for SRLV, treated with the ethyl acetate fraction of M. azedarach (EAF-MA) for three days, while the other group consumed milk from does also carrying the virus with the respective extract twice a day for five days. After undergoing treatment, all animals began to receive thermized milk until weaning (60 days) and were monitored for six months using nested polymerase chain reaction (nPCR) and western blot (WB) tests. The study revealed cumulative percentages of positive animals in WB or nPCR in the milk group of 66.66% on the seventh day, 83.33% in the following week, and 100% at 120 days, while the colostrum group showed values of 66.66% at 14 days, 83.33% at 90 days, and 100% at 120 days. Variation and intermittency were observed in viral detection, but all animals tested positive in WB or nPCR at some point. A potential delay in infection was observed, which was more significant in the colostrum group. The need for the combination of serological and molecular tests for a more efficient detection of the disease is also emphasized.
Collapse
Affiliation(s)
| | | | | | | | | | - Ana Milena César Lima
- Regional Scientific Development Fellowship of the National Council for Scientific and Technological Development (DCR-CNPq/FUNCAP), Level C, Embrapa Goats & Sheep, Sobral, Ceará, Brazil
| | | | | | - Gabriel Paula Amaral
- Graduate Program in Animal Science, Vale Do Acaraú State University, Sobral, Ceará, Brazil
| | | |
Collapse
|
15
|
Zeng YC, Young OJ, Si L, Ku MW, Isinelli G, Rajwar A, Jiang A, Wintersinger CM, Graveline AR, Vernet A, Sanchez M, Ryu JH, Kwon IC, Goyal G, Ingber DE, Shih WM. DNA origami vaccine (DoriVac) nanoparticles improve both humoral and cellular immune responses to infectious diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.29.573647. [PMID: 38260393 PMCID: PMC10802255 DOI: 10.1101/2023.12.29.573647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Current SARS-CoV-2 vaccines have demonstrated robust induction of neutralizing antibodies and CD4+ T cell activation, however CD8+ responses are variable, and the duration of immunity and protection against variants are limited. Here we repurposed our DNA origami vaccine platform, DoriVac, for targeting infectious viruses, namely SARS-CoV-2, HIV, and Ebola. The DNA origami nanoparticle, conjugated with infectious-disease-specific HR2 peptides, which act as highly conserved antigens, and CpG adjuvant at precise nanoscale spacing, induced neutralizing antibodies, Th1 CD4+ T cells, and CD8+ T cells in naïve mice, with significant improvement over a bolus control. Pre-clinical studies using lymph-node-on-a-chip systems validated that DoriVac, when conjugated with antigenic peptides or proteins, induced promising cellular immune responses in human cells. These results suggest that DoriVac holds potential as a versatile, modular vaccine platform, capable of inducing both humoral and cellular immunities. The programmability of this platform underscores its potential utility in addressing future pandemics.
Collapse
Affiliation(s)
- Yang C. Zeng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Olivia J. Young
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Longlong Si
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Min Wen Ku
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Giorgia Isinelli
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Anjali Rajwar
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Amanda Jiang
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Chris M. Wintersinger
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Amanda R. Graveline
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Andyna Vernet
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Melinda Sanchez
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Ju Hee Ryu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA
| | - William M. Shih
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
16
|
Luo G, Aldridge K, Chen T, Aslot V, Kim BG, Han EH, Singh N, Li S, Xiao TS, Sporn MB, Letterio JJ. The synthetic oleanane triterpenoid CDDO-2P-Im binds GRP78/BiP to induce unfolded protein response-mediated apoptosis in myeloma. Mol Oncol 2023; 17:2526-2545. [PMID: 37149844 DOI: 10.1002/1878-0261.13447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/20/2023] [Accepted: 05/05/2023] [Indexed: 05/09/2023] Open
Abstract
Synthetic oleanane triterpenoids (SOTs) are small molecules with broad anticancer properties. A recently developed SOT, 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4(-pyridin-2-yl)-1H-imidazole (CDDO-2P-Im or '2P-Im'), exhibits enhanced activity and improved pharmacokinetics over CDDO-Im, a previous generation SOT. However, the mechanisms leading to these properties are not defined. Here, we show the synergy of 2P-Im and the proteasome inhibitor ixazomib in human multiple myeloma (MM) cells and 2P-Im activity in a murine model of plasmacytoma. RNA sequencing and quantitative reverse transcription PCR revealed the upregulation of the unfolded protein response (UPR) in MM cells upon 2P-lm treatment, implicating the activation of the UPR as a key step in 2P-Im-induced apoptosis. Supporting this hypothesis, the deletion of genes encoding either protein kinase R-like endoplasmic reticulum kinase (PERK) or DNA damage-inducible transcript 3 protein (DDIT3; also known as CHOP) impaired the MM response to 2P-Im, as did treatment with ISRIB, integrated stress response inhibitor, which inhibits UPR signaling downstream of PERK. Finally, both drug affinity responsive target stability and thermal shift assays demonstrated direct binding of 2P-Im to endoplasmic reticulum chaperone BiP (GRP78/BiP), a stress-inducible key signaling molecule of the UPR. These data reveal GRP78/BiP as a novel target of SOTs, and specifically of 2P-Im, and suggest the potential broader utility of this class of small molecules as modulators of the UPR.
Collapse
Affiliation(s)
- George Luo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Toby Chen
- Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Vivek Aslot
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Byung-Gyu Kim
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- The Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Eun Hyang Han
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- The Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Neelima Singh
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- The Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Sai Li
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Tsan Sam Xiao
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - John J Letterio
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- The Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
17
|
Al Ibrahim M, Akissi ZLE, Desmarets L, Lefèvre G, Samaillie J, Raczkiewicz I, Sahpaz S, Dubuisson J, Belouzard S, Rivière C, Séron K. Discovery of Anti-Coronavirus Cinnamoyl Triterpenoids Isolated from Hippophae rhamnoides during a Screening of Halophytes from the North Sea and Channel Coasts in Northern France. Int J Mol Sci 2023; 24:16617. [PMID: 38068938 PMCID: PMC10705938 DOI: 10.3390/ijms242316617] [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: 10/31/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
The limited availability of antiviral therapy for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spurred the search for novel antiviral drugs. Here, we investigated the potential antiviral properties of plants adapted to high-salt environments collected in the north of France. Twenty-five crude methanolic extracts obtained from twenty-two plant species were evaluated for their cytotoxicity and antiviral effectiveness against coronaviruses HCoV-229E and SARS-CoV-2. Then, a bioguided fractionation approach was employed. The most active crude methanolic extracts were partitioned into three different sub-extracts. Notably, the dichloromethane sub-extract of the whole plant Hippophae rhamnoides L. demonstrated the highest antiviral activity against both viruses. Its chemical composition was evaluated by ultra-high performance liquid chromatography (UHPLC) coupled with mass spectrometry (MS) and then it was fractionated by centrifugal partition chromatography (CPC). Six cinnamoyl triterpenoid compounds were isolated from the three most active fractions by preparative high-performance liquid chromatography (HPLC) and identified by high resolution MS (HR-MS) and mono- and bi-dimensional nuclear magnetic resonance (NMR). Specifically, these compounds were identified as 2-O-trans-p-coumaroyl-maslinic acid, 3β-hydroxy-2α-trans-p-coumaryloxy-urs-12-en-28-oic acid, 3β-hydroxy-2α-cis-p-coumaryloxy-urs-12-en-28-oic acid, 3-O-trans-caffeoyl oleanolic acid, a mixture of 3-O-trans-caffeoyl oleanolic acid/3-O-cis-caffeoyl oleanolic acid (70/30), and 3-O-trans-p-coumaroyl oleanolic acid. Infection tests demonstrated a dose-dependent inhibition of these triterpenes against HCoV-229E and SARS-CoV-2. Notably, cinnamoyl oleanolic acids displayed activity against both SARS-CoV-2 and HCoV-229E. Our findings suggest that Hippophae rhamnoides could represent a source of potential antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Malak Al Ibrahim
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR9017—Center for Infection and Immunity of Lille (CIIL), F-59000 Lille, France; (M.A.I.); (L.D.); (I.R.); (J.D.); (S.B.)
- BioEcoAgro, Joint Research Unit 1158, University of Lille, INRAE, University of. Liège, UPJV, YNCREA, University of Artois, University Littoral Côte d’Opale, ICV—Institut Charles Viollette, F-59650 Villeneuve d’Ascq, France; (Z.L.E.A.); (G.L.); (J.S.); (S.S.)
| | - Zachee Louis Evariste Akissi
- BioEcoAgro, Joint Research Unit 1158, University of Lille, INRAE, University of. Liège, UPJV, YNCREA, University of Artois, University Littoral Côte d’Opale, ICV—Institut Charles Viollette, F-59650 Villeneuve d’Ascq, France; (Z.L.E.A.); (G.L.); (J.S.); (S.S.)
| | - Lowiese Desmarets
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR9017—Center for Infection and Immunity of Lille (CIIL), F-59000 Lille, France; (M.A.I.); (L.D.); (I.R.); (J.D.); (S.B.)
| | - Gabriel Lefèvre
- BioEcoAgro, Joint Research Unit 1158, University of Lille, INRAE, University of. Liège, UPJV, YNCREA, University of Artois, University Littoral Côte d’Opale, ICV—Institut Charles Viollette, F-59650 Villeneuve d’Ascq, France; (Z.L.E.A.); (G.L.); (J.S.); (S.S.)
| | - Jennifer Samaillie
- BioEcoAgro, Joint Research Unit 1158, University of Lille, INRAE, University of. Liège, UPJV, YNCREA, University of Artois, University Littoral Côte d’Opale, ICV—Institut Charles Viollette, F-59650 Villeneuve d’Ascq, France; (Z.L.E.A.); (G.L.); (J.S.); (S.S.)
| | - Imelda Raczkiewicz
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR9017—Center for Infection and Immunity of Lille (CIIL), F-59000 Lille, France; (M.A.I.); (L.D.); (I.R.); (J.D.); (S.B.)
| | - Sevser Sahpaz
- BioEcoAgro, Joint Research Unit 1158, University of Lille, INRAE, University of. Liège, UPJV, YNCREA, University of Artois, University Littoral Côte d’Opale, ICV—Institut Charles Viollette, F-59650 Villeneuve d’Ascq, France; (Z.L.E.A.); (G.L.); (J.S.); (S.S.)
| | - Jean Dubuisson
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR9017—Center for Infection and Immunity of Lille (CIIL), F-59000 Lille, France; (M.A.I.); (L.D.); (I.R.); (J.D.); (S.B.)
| | - Sandrine Belouzard
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR9017—Center for Infection and Immunity of Lille (CIIL), F-59000 Lille, France; (M.A.I.); (L.D.); (I.R.); (J.D.); (S.B.)
| | - Céline Rivière
- BioEcoAgro, Joint Research Unit 1158, University of Lille, INRAE, University of. Liège, UPJV, YNCREA, University of Artois, University Littoral Côte d’Opale, ICV—Institut Charles Viollette, F-59650 Villeneuve d’Ascq, France; (Z.L.E.A.); (G.L.); (J.S.); (S.S.)
| | - Karin Séron
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR9017—Center for Infection and Immunity of Lille (CIIL), F-59000 Lille, France; (M.A.I.); (L.D.); (I.R.); (J.D.); (S.B.)
| |
Collapse
|
18
|
Gao J, Cao C, Shi M, Hong S, Guo S, Li J, Liang T, Song P, Xu R, Li N. Kaempferol inhibits SARS-CoV-2 invasion by impairing heptad repeats-mediated viral fusion. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154942. [PMID: 37421767 PMCID: PMC10289257 DOI: 10.1016/j.phymed.2023.154942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND The continuous evolution of SARS-CoV-2 has underscored the development of broad-spectrum prophylaxis. Antivirals targeting the membrane fusion process represent promising paradigms. Kaempferol (Kae), an ubiquitous plant flavonol, has been shown efficacy against various enveloped viruses. However, its potential in anti-SARS-CoV-2 invasion remains obscure. PURPOSE To evaluate capabilities and mechanisms of Kae in preventing SARS-CoV-2 invasion. METHODS To avoid interference of viral replication, virus-like particles (VLPs) constructed with luciferase reporter were applied. To investigate the antiviral potency of Kae, human induced pluripotent stem cells (hiPSC)-derived alveolar epithelial cells type II (AECII) and human ACE2 (hACE2) transgenic mice were utilized as in vitro and in vivo models, respectively. Using dual split protein (DSP) assays, inhibitory activities of Kae in viral fusion were determined in Alpha, Delta and Omicron variants of SARS-CoV-2, as well as in SARS-CoV and MERS-CoV. To further reveal molecular determinants of Kae in restricting viral fusion, synthetic peptides corresponding to the conserved heptad repeat (HR) 1 and 2, involved in viral fusion, and the mutant form of HR2 were explored by circular dichroism and native polyacrylamide gel electrophoresis. RESULTS Kae inhibited SARS-CoV-2 invasion both in vitro and in vivo, which was mainly attributed to its suppressive effects on viral fusion, but not endocytosis, two pathways that mediate viral invasion. In accordance with the proposed model of anti-fusion prophylaxis, Kae functioned as a pan-inhibitor of viral fusion, including three emerged highly pathogenic coronaviruses, and the currently circulating Omicron BQ.1.1 and XBB.1 variants of SARS-CoV-2. Consistent with the typical target of viral fusion inhibitors, Kae interacted with HR regions of SARS-CoV-2 S2 subunits. Distinct from previous inhibitory fusion peptides which prevent the formation of six-helix bundle (6-HB) by competitively interacting with HRs, Kae deformed HR1 and directly reacted with lysine residues within HR2 region, the latter of which was considered critical for the preservation of stabilized S2 during SARS-CoV-2 invasion. CONCLUSIONS Kae prevents SARS-CoV-2 infection by blocking membrane fusion and possesses a broad-spectrum anti-fusion ability. These findings provide valuable insights into potential benefits of Kae-containing botanical products as a complementary prophylaxis, especially during the waves of breakthrough infections and re-infections.
Collapse
Affiliation(s)
- Junwei Gao
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Can Cao
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Mingfei Shi
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shihao Hong
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shijie Guo
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jing Li
- Department of Nephropathy, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Tengxiao Liang
- Department of Emergency, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Ping Song
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China..
| | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China..
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China..
| |
Collapse
|
19
|
Li C, Zhang J, Wu Q, Kumar A, Pan G, Kelvin DJ. Nifuroxazide Activates the Parthanatos to Overcome TMPRSS2:ERG Fusion-Positive Prostate Cancer. Mol Cancer Ther 2023; 22:306-316. [PMID: 36622760 PMCID: PMC9978883 DOI: 10.1158/1535-7163.mct-22-0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/24/2022] [Accepted: 12/30/2022] [Indexed: 01/10/2023]
Abstract
Fusion of the E-26 transformation-specific (ETS)-related gene (ERG) with transmembrane serine protease 2 (TMPRSS2) is a crucial step in the occurrence and progression of approximately 50% of prostate cancers. Despite significant progress in drug discovery, ERG inhibitors have yet to be approved for the clinical treatment of prostate cancer. In this study, we used computer-aided drug design (CADD)-based virtual screening to screen for potential inhibitors of ERG. In vivo and in vitro methods revealed that nifuroxazide (NFZ) inhibited the proliferation of a TMPRSS2:ERG fusion-positive prostate cancer cell line (VCaP) with an IC50 lower than that of ERG-negative prostate cancer cell lines (LNCaP, DU145, and WPMY cells). Poly [ADP-ribose] polymerase 1, the critical mediator of parthanatos, is known to bind ERG and is required for ERG-mediated transcription. NFZ blocked this interaction and overly activated PARP1, leading to cell death that was reduced by olaparib, a PARP1 inhibitor. These results show that NFZ inhibits ERG, leading to parthanatic cell death.
Collapse
Affiliation(s)
- Chengxun Li
- Laboratory of Immunity, Shantou University Medical College, Guangdong, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jiale Zhang
- Laboratory of Immunity, Shantou University Medical College, Guangdong, China
| | - Qiming Wu
- Laboratory of Immunity, Shantou University Medical College, Guangdong, China
| | - Anuj Kumar
- Laboratory of Immunity, Shantou University Medical College, Guangdong, China
- Department of Microbiology and Immunology, Faculty of Medicine, Canadian Center for Vaccinology, Dalhousie University, Halifax, NS, Canada
| | - Guihong Pan
- Laboratory of Immunity, Shantou University Medical College, Guangdong, China
| | - David J. Kelvin
- Laboratory of Immunity, Shantou University Medical College, Guangdong, China
- Department of Microbiology and Immunology, Faculty of Medicine, Canadian Center for Vaccinology, Dalhousie University, Halifax, NS, Canada
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Guangdong, China
- Corresponding Author: David J. Kelvin, Laboratory of Immunity, Shantou University Medical College, Guangdong, China. E-mail:
| |
Collapse
|
20
|
Liu M, Wang J, Wan X, Li B, Guan M, Ning X, Hu X, Li S, Liu S, Song G. Discovery and structural optimization of 3-O-β-Chacotriosyl betulonic acid saponins as potent fusion inhibitors of Omicron virus infections. Bioorg Chem 2023; 131:106316. [PMID: 36508939 PMCID: PMC9729598 DOI: 10.1016/j.bioorg.2022.106316] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/07/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
The recent global Omicron epidemics underscore the great need for the development of small molecule therapeutics with appropriate mechanisms. The trimeric spike protein (S) of SARS-CoV-2 plays a pivotal role in mediating viral entry into host cells. We continued our efforts to develop small-molecule SARS-CoV-2 entry inhibitors. In this work, two sets of BA derivatives were designed and synthesized based on the hit BA-1 that was identified as a novel SARS-CoV-2 entry inhibitor. Compound BA-4, the most potent one, showed broad inhibitory activities against pOmicron and other pseudotyped variants with EC50 values ranging 2.73 to 5.19 μM. Moreover, pSARS-CoV-2 assay, SPR analysis, Co-IP assay and the cell-cell fusion assay coupled with docking and mutagenesis studies revealed that BA-4 could stabilize S in the pre-fusion step to interfere with the membrane fusion, thereby displaying promising inhibition against Omicron entry.
Collapse
Affiliation(s)
- Mingjian Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Jinshen Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Wan
- Huizhou Health Sciences Polytechnic, Huizhou 516025, China
| | - Baixi Li
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Mingming Guan
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyun Ning
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Xiaojie Hu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Sumei Li
- Department of Human anatomy, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou 510515, China.
| | - Gaopeng Song
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
21
|
Anti-HIV Potential of Beesioside I Derivatives as Maturation Inhibitors: Synthesis, 3D-QSAR, Molecular Docking and Molecular Dynamics Simulations. Int J Mol Sci 2023; 24:ijms24021430. [PMID: 36674943 PMCID: PMC9867151 DOI: 10.3390/ijms24021430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
HIV-1 maturation is the final step in the retroviral lifecycle that is regulated by the proteolytic cleavage of the Gag precursor protein. As a first-in-class HIV-1 maturation inhibitor (MI), bevirimat blocks virion maturation by disrupting capsid-spacer peptide 1 (CA-SP1) cleavage, which acts as the target of MIs. Previous alterations of beesioside I (1) produced (20S,24S)-15ꞵ,16ꞵ-diacetoxy-18,24; 20,24-diepoxy-9,19-cyclolanostane-3ꞵ,25-diol 3-O-3′,3′-dimethylsuccinate (3, DSC), showing similar anti-HIV potency compared to bevirimat. To ascertain the binding modes of this derivative, further modification of compound 1 was conducted. Three-dimensional quantitative structure−activity relationship (3D-QSAR) analysis combined with docking simulations and molecular dynamics (MD) were conducted. Five new derivatives were synthesized, among which compound 3b showed significant activity against HIV-1NL4-3 with an EC50 value of 0.28 µM. The developed 3D-QSAR model resulted in great predictive ability with training set (r2 = 0.99, q2 = 0.55). Molecular docking studies were complementary to the 3D-QSAR analysis, showing that DSC was differently bound to CA-SP1 with higher affinity than that of bevirimat. MD studies revealed that the complex of the ligand and the protein was stable, with root mean square deviation (RMSD) values <2.5 Å. The above results provided valuable insights into the potential of DSC as a prototype to develop new antiviral agents.
Collapse
|
22
|
Qi JH, Chen PY, Cai DY, Wang Y, Wei YL, He SP, Zhou W. Exploring novel targets of sitagliptin for type 2 diabetes mellitus: Network pharmacology, molecular docking, molecular dynamics simulation, and SPR approaches. Front Endocrinol (Lausanne) 2023; 13:1096655. [PMID: 36699034 PMCID: PMC9868454 DOI: 10.3389/fendo.2022.1096655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Diabetes has become a serious global public health problem. With the increasing prevalence of type 2 diabetes mellitus (T2DM), the incidence of complications of T2DM is also on the rise. Sitagliptin, as a targeted drug of DPP4, has good therapeutic effect for T2DM. It is well known that sitagliptin can specifically inhibit the activity of DPP4 to promote insulin secretion, inhibit islet β cell apoptosis and reduce blood glucose levels, while other pharmacological mechanisms are still unclear, such as improving insulin resistance, anti-inflammatory, anti-oxidative stress, and anti-fibrosis. The aim of this study was to explore novel targets and potential signaling pathways of sitagliptin for T2DM. METHODS Firstly, network pharmacology was applied to find the novel target most closely related to DPP4. Semi-flexible molecular docking was performed to confirm the binding ability between sitagliptin and the novel target, and molecular dynamics simulation (MD) was carried to verify the stability of the complex formed by sitagliptin and the novel target. Furthermore, surface-plasmon resonance (SPR) was used to explored the affinity and kinetic characteristics of sitagliptin with the novel target. Finally, the molecular mechanism of sitagliptin for T2DM was predicted by the enrichment analysis of GO function and KEGG pathway. RESULTS In this study, we found the cell surface receptor-angiotensin-converting enzyme 2 (ACE2) most closely related to DPP4. Then, we confirmed that sitagliptin had strong binding ability with ACE2 from a static perspective, and the stability of sitagliptin-ACE2 complex had better stability and longer binding time than BAR708-ACE2 in simulated aqueous solution within 50 ns. Significantly, we have demonstrated a strong affinity between sitagliptin and ACE2 on SPR biosensor, and their kinetic characteristics were "fast binding/fast dissociation". The guiding significance of clinical administration: low dose can reach saturation, but repeated administration was needed. Finally, there was certain relationship between COVID-19 and T2DM, and ACE2/Ang-(1-7)/Mas receptor (MasR) axis may be the important pathway of sitagliptin targeting ACE2 for T2DM. CONCLUSION This study used different methods to prove that ACE2 may be another novel target of sitagliptin for T2DM, which extended the application of ACE2 in improving diabetes mellitus.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
23
|
Guo Y, Ma A, Wang X, Yang C, Chen X, Li G, Qiu F. Research progress on the antiviral activities of natural products and their derivatives: Structure–activity relationships. Front Chem 2022; 10:1005360. [PMID: 36311429 PMCID: PMC9596788 DOI: 10.3389/fchem.2022.1005360] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022] Open
Abstract
Viruses spread rapidly and are well-adapted to changing environmental events. They can infect the human body readily and trigger fatal diseases. A limited number of drugs are available for specific viral diseases, which can lead to non-efficacy against viral variants and drug resistance, so drugs with broad-spectrum antiviral activity are lacking. In recent years, a steady stream of new viral diseases has emerged, which has prompted development of new antiviral drugs. Natural products could be employed to develop new antiviral drugs because of their innovative structures and broad antiviral activities. This review summarizes the progress of natural products in antiviral research and their bright performance in drug resistance issues over the past 2 decades. Moreover, it fully discusses the effect of different structural types of natural products on antiviral activity in terms of structure–activity relationships. This review could provide a foundation for the development of antiviral drugs.
Collapse
Affiliation(s)
- Yajing Guo
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Anna Ma
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinyan Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chen Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xi Chen
- School of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xi Chen, ; Gen Li,
| | - Gen Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xi Chen, ; Gen Li,
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjfin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
24
|
A novel cyclic γ-AApeptide-based long-acting pan-coronavirus fusion inhibitor with potential oral bioavailability by targeting two sites in spike protein. Cell Discov 2022; 8:88. [PMID: 36075899 PMCID: PMC9453727 DOI: 10.1038/s41421-022-00455-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/31/2022] [Indexed: 12/13/2022] Open
Abstract
The receptor-binding domain (RBD) in S1 subunit and heptad repeat 1 (HR1) domain in S2 subunit of SARS-CoV-2 spike (S) protein are the targets of neutralizing antibodies (nAbs) and pan-coronavirus (CoV) fusion inhibitory peptides, respectively. However, neither nAb- nor peptide-based drugs can be used orally. In this study, we screened a one-bead-two-compound (OBTC) cyclic γ-AApeptide library against SARS-CoV-2 S protein and identified a hit: S-20 with potent membrane fusion inhibitory activity, but moderate selectivity index (SI). After modification, one derivative, S-20-1, exhibited improved fusion inhibitory activity and SI (>1000). S-20-1 could effectively inhibit infection by pseudotyped and authentic SARS-CoV-2 and pseudotyped variants of concern (VOCs), including B.1.617.2 (Delta) and B.1.1.529 (Omicron), as well as MERS-CoV, SARS-CoV, HCoV-OC43, HCoV-229E, and HCoV-NL63. It could also inhibit infection of a pseudotyped SARS-related coronavirus WIV1 (SARSr-CoV-WIV1) from bats. Intranasal application of S-20-1 to mice before or after challenge with HCoV-OC43 or SARS-CoV-2 provided significant protection from infection. Importantly, S-20-1 was highly resistant to proteolytic degradation, had long half-life, and possessed favorable oral bioavailability. Mechanistic studies suggest that S-20-1 binds with high affinity to RBD in S1 and HR1 domain in S2 of SARS-CoV-2 S protein. Thus, with its pan-CoV fusion and entry inhibitory activity by targeting two sites in S protein, desirable half-life, and promising oral bioavailability, S-20-1 is a potential candidate for further development as a novel therapeutic and prophylactic drug against infection by SARS-CoV-2 and its variants, as well as future emerging and reemerging CoVs.
Collapse
|
25
|
Abietic, maleopimaric and quinopimaric dipeptide Ugi-4CR derivatives and their potency against influenza A and SARS-CoV-2. Nat Prod Res 2022; 37:1954-1960. [PMID: 35975755 DOI: 10.1080/14786419.2022.2112040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A set of 12 abietane diterpene derivatives have been synthesised by the Ugi-four component reaction (Ugi-4CR) and tested for cytotoxicity and activity against influenza virus A/Puerto Rico/8/34 (H1N1) and SARS-CoV-2 pseudovirus. Five dipeptide derivatives demonstrated a selectivity index (SI) higher than 10 and IC50 values from 2 to 32 μM against influenza virus. Compound 11 was found to be a lead with SI of 200, and time-of-addition experiments showed the viral entry into the cell and the binding of the virus to the receptor as a possible target. Compound 7 was the only one showed weak anti-SARS-CoV-2 activity with EC50 value of 80.96 µM. Taken together, our data suggest the potency of diterpene acids-Ugi products as new effective anti-influenza compounds.
Collapse
|
26
|
Han S, Li H, Chen W, Yang L, Tong X, Zuo J, Hu Y. Discovery of potent ebola entry inhibitors with (3S,4aS,8aS)-2-(3-amino-2-hydroxypropyl) decahydroisoquinoline-3-carboxamide scaffold. Eur J Med Chem 2022; 240:114608. [PMID: 35872393 DOI: 10.1016/j.ejmech.2022.114608] [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: 06/07/2022] [Revised: 07/03/2022] [Accepted: 07/10/2022] [Indexed: 11/15/2022]
Abstract
Ebola virus (EBOV), one member of the family Filoviridae, can causes hemorrhagic fever and other severe diseases in humans with a high mortality rate (25-90%). Until recently, there were no approved drugs and very limited treatment method for Ebola virus disease. In this study, we discovered a series of potent Ebola entry inhibitors with the (3S,4aS,8aS)-2-(3-amino-2-hydroxypropyl)decahydroisoquinoline-3-carboxamide scaffold from high-throughput screening in reported pseudotyped virus system. Further optimization resulted a most potent compound 28 (IC50= 0.05 μM, SI = 98), which displayed 3-fold potency compared to the known inhibitor Toremifene (IC50= 0.17 μM, SI = 55). Moreover, compound 28 exhibited the remarkable selectivity between EBOV-GP and VSV-G (Spec. Index = 58), thus could exclude nonspecific effects. Structure-activity relationship and molecular docking analysis of the new chemical scaffold provided more information on the binding modes and the spare volume at the binding cavity, thus can guide the design of the further potent compounds.
Collapse
Affiliation(s)
- Sheng Han
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Heng Li
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weixiong Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Yang
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiankun Tong
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jianping Zuo
- Immunological Disease Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Youhong Hu
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, 1 Xiangshanzhi Road, Hangzhou, 310024, China.
| |
Collapse
|
27
|
Wang X, Chen Y, Shi H, Zou P. Erythromycin Estolate Is a Potent Inhibitor Against HCoV-OC43 by Directly Inactivating the Virus Particle. Front Cell Infect Microbiol 2022; 12:905248. [PMID: 35873167 PMCID: PMC9301004 DOI: 10.3389/fcimb.2022.905248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/10/2022] [Indexed: 12/22/2022] Open
Abstract
In addition to antibacterial effects, macrolide antibiotics exhibit other extensive pharmacological effects, such as anti-inflammatory and antiviral activities. Erythromycin estolate, one of the macrolide antibiotics, was previously investigated to effectively inhibit infections of various flaviviruses including Zika virus, dengue virus, and yellow fever virus, but its antiviral effect against human coronavirus remains unknown. Thus, the current study was designed to evaluate the antiviral efficacy of erythromycin estolate against human coronavirus strain OC43 (HCoV-OC43) and to illustrate the underlying mechanisms. Erythromycin estolate effectively inhibited HCoV-OC43 infection in different cell types and significantly reduced virus titers at safe concentration without cell cytotoxicity. Furthermore, erythromycin estolate was identified to inhibit HCoV-OC43 infection at the early stage and to irreversibly inactivate virus by disrupting the integrity of the viral membrane whose lipid component might be the target of action. Together, it was demonstrated that erythromycin estolate could be a potential therapeutic drug for HCoV-OC43 infection.
Collapse
Affiliation(s)
- Xiaohuan Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongkang Chen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Huichun Shi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Peng Zou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Peng Zou,
| |
Collapse
|
28
|
Zhou J, Rong XL, Cao X, Tang Q, Liu D, Jin YH, Shi XX, Zhong M, Zhao Y, Yang Y. Assembly of Poly(ethylene glycol)ylated Oleanolic Acid on a Linear Polymer as a Pseudomucin for Influenza Virus Inhibition and Adsorption. Biomacromolecules 2022; 23:3213-3221. [PMID: 35797332 DOI: 10.1021/acs.biomac.2c00314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Biomimicry of the mucin barrier function is an efficient strategy to counteract influenza. We report the simple aminolyzation of poly(methyl vinyl ether-alt-maleic anhydride) (PM) using amine-terminated poly(ethylene glycol)ylated oleanolic acid (OAPEG) to mimic the mucin structure and its adsorption of the influenza virus. Direct interactions between influenza hemagglutinin (HA) and the prepared macromolecule evaluated by surface plasmon resonance and isothermal titration calorimetry demonstrated that the multivalent presentation of OAPEG on PM enhanced the binding affinity to HA with a decrease in KD of approximately three orders of magnitude compared with monomeric OAPEG. Moreover, hemagglutination inhibition assay, viral growth inhibition assay, and cytopathic effect reduction assay indicated that the nonglycosylated polymer could mimic natural heavily glycosylated mucin and thus promote the attachment of the virus in a subnanomolar range. Further investigation of the antiviral effects via time-of-addition assay, dynamic light scattering experiments, and transmission electron microscopy photographs indicated that the pseudomucin could adsorb the virion particles and synergistically inhibit the early attachment and final release steps of the influenza infection cycle. These findings demonstrate the effectiveness of the macromolecule in the physical sequestration and prevention of viral infection. Notably, due to its structural similarities with mucin, the biomacropolymer also has the potential for the rational design of antiviral drugs, influenza adsorbents, or filtration materials and the construction of model systems to explore protection against other pathogenic viruses.
Collapse
Affiliation(s)
- JiaPing Zhou
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China.,Research Centre of Modern Analytical Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xue-Lin Rong
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xuan Cao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Qi Tang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Dong Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Yin-Hua Jin
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xiao-Xiao Shi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Ming Zhong
- Medical College of Shaoguan University, Shaoguan, Guangdong Province 512026, China
| | - YueTao Zhao
- School of Life Sciences, Central South University, Changsha, Hunan Province 410013, China
| | - Yang Yang
- Research Centre of Modern Analytical Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China.,China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| |
Collapse
|
29
|
Gao Y, Tai W, Wang X, Jiang S, Debnath AK, Du L, Chen S. A gossypol derivative effectively protects against Zika and dengue virus infection without toxicity. BMC Biol 2022; 20:143. [PMID: 35706035 PMCID: PMC9202104 DOI: 10.1186/s12915-022-01344-w] [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: 12/12/2020] [Accepted: 06/07/2022] [Indexed: 11/28/2022] Open
Abstract
Background Zika virus (ZIKV) and dengue virus (DENV) cause microcephaly and dengue hemorrhagic fever, respectively, leading to severe problems. No effective antiviral agents are approved against infections of these flaviviruses, calling for the need to develop potent therapeutics. We previously identified gossypol as an effective inhibitor against ZIKV and DENV infections, but this compound is toxic and not suitable for in vivo treatment. Results In this study, we showed that gossypol derivative ST087010 exhibited potent and broad-spectrum in vitro inhibitory activity against infections of at least ten ZIKV strains isolated from different hosts, time periods, and countries, as well as DENV-1-4 serotypes, and significantly reduced cytotoxicity compared to gossypol. It presented broad-spectrum in vivo protective efficacy, protecting ZIKV-infected Ifnar1−/− mice from lethal challenge, with increased survival and reduced weight loss. Ifnar1−/− mice treated with this gossypol derivative decreased viral titers in various tissues, including the brain and testis, after infection with ZIKV at different human isolates. Moreover, ST087010 potently blocked ZIKV vertical transmission in pregnant Ifnar1−/− mice, preventing ZIKV-caused fetal death, and it was safe for pregnant mice and their pups. It also protected DENV-2-challenged Ifnar1−/− mice against viral replication by reducing the viral titers in the brain, kidney, heart, and sera. Conclusions Overall, our data indicate the potential for further development of this gossypol derivative as an effective and safe broad-spectrum therapeutic agent to treat ZIKV and DENV diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01344-w.
Collapse
Affiliation(s)
- Yaning Gao
- Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Wanbo Tai
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Xinyi Wang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.,Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Asim K Debnath
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.
| | - Shizhong Chen
- Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
30
|
Li H, Wang S, Ma W, Cheng B, Yi Y, Ma X, Xiao S, Zhang L, Zhou D. Discovery of Pentacyclic Triterpenoid PROTACs as a Class of Effective Hemagglutinin Protein Degraders. J Med Chem 2022; 65:7154-7169. [PMID: 35579113 DOI: 10.1021/acs.jmedchem.1c02013] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Influenza hemagglutinin that drives viral entry into cells via the membrane fusion process is an up-and-coming antiviral drug target. Herein, we described for the first time the design, synthesis, and biological characteristics of a new class of pentacyclic triterpenoid-based proteolysis targeting chimeras (PROTACs) to enhance the degradation of hemagglutinin target. Among these PROTACs, V3 showed the best degradation effect on the hemagglutinin with a median degradation concentration of 1.44 μM in a ubiquitin and proteasome-dependent manner and broad-spectrum anti-influenza A virus activity but not affected the entry of influenza virus. Moreover, intravenous injection of V3 protected mice against influenza A virus-induced toxic effects. Further diazirine-containing photo-crosslinking mass spectrometric analysis of hemagglutinin complexes indicated crosslinking to Asn15, Thr31, and Asn27, a novel target of hemagglutinin. Taken together, our data revealed that oleanolic acid-based PROTACs could degrade hemagglutinin protein, providing a new direction toward the discovery of potential anti-influenza drugs.
Collapse
Affiliation(s)
- Haiwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shouxin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenxiao Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Boyang Cheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yanliang Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinyuan Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Sulong Xiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.,Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China.,Ningbo Institute of Marine Medicine, Peking University, Ningbo 315010, China
| |
Collapse
|
31
|
“Malancha” [Alternanthera philoxeroides (Mart.) Griseb.]: A Potential Therapeutic Option against Viral Diseases. Biomolecules 2022; 12:biom12040582. [PMID: 35454170 PMCID: PMC9025398 DOI: 10.3390/biom12040582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Alternanthera philoxeroides (Mart.) Griseb., commonly known as “Alligator weed” in English, and “Malancha” in Bengali, is a leafy vegetable from the family Amaranthaceae A. L. de Jussieu. This species is native to China, particularly to the provinces around the Yangtze River, other Far East and South-East Asian countries, and countries from other continents (e.g., South America). This plant also grows in certain areas in Australia, New Zealand, and the USA. While in Bangladesh the leaves of this plant are consumed as a vegetable, in China, this plant has been used widely as a traditional remedy for the treatment of various viral diseases (e.g., measles, influenza, and haemorrhagic fever). Flavonoids and saponins are the two largest groups of phytochemicals produced by this plant, and the antiviral property of this plant and its compounds has been studied extensively. This review article reviews all published literature on this plant and critically appraises its phytochemical profile linking to biomolecular interactions and therapeutic potential, particularly, against viral diseases.
Collapse
|
32
|
Liang S, Ma X, Li M, Yi Y, Gao Q, Zhang Y, Zhang L, Zhou D, Xiao S. Novel β-Cyclodextrin-Based Heptavalent Glycyrrhetinic Acid Conjugates: Synthesis, Characterization, and Anti-Influenza Activity. Front Chem 2022; 10:836955. [PMID: 35494649 PMCID: PMC9039011 DOI: 10.3389/fchem.2022.836955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
In our continuing efforts toward the design of novel pentacyclic triterpene derivatives as potential anti-influenza virus entry inhibitors, a series of homogeneous heptavalent glycyrrhetinic acid derivatives based on β-cyclodextrin scaffold were designed and synthesized by click chemistry. The structure was unambiguously characterized by NMR, IR, and MALDI-TOF-MS measurements. Seven conjugates showed sufficient inhibitory activity against influenza virus infection based on the cytopathic effect reduction assay with IC50 values in the micromolar range. The interactions of conjugate 37, the most potent compound (IC50 = 2.86 μM, CC50 > 100 μM), with the influenza virus were investigated using the hemagglutination inhibition assay. Moreover, the surface plasmon resonance assay further confirmed that compound 37 bound to the influenza HA protein specifically with a dissociation constant of 5.15 × 10−7 M. Our results suggest the promising role of β-cyclodextrin as a scaffold for preparing a variety of multivalent compounds as influenza entry inhibitors.
Collapse
Affiliation(s)
- Shuobin Liang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xinyuan Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Man Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yanliang Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qianqian Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yongmin Zhang
- Sorbonne Université, Institut Parisien de Chimie Moléculaire, CNRS UMR 8232, Paris, France
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo, China
| | - Sulong Xiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- *Correspondence: Sulong Xiao,
| |
Collapse
|
33
|
Gomaa AA, Mohamed HS, Abd-Ellatief RB, Gomaa MA, Hammam DS. Advancing combination treatment with glycyrrhizin and boswellic acids for hospitalized patients with moderate COVID-19 infection: a randomized clinical trial. Inflammopharmacology 2022; 30:477-486. [PMID: 35233748 PMCID: PMC8886861 DOI: 10.1007/s10787-022-00939-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Recent evidence points to a potential therapeutic role for glycyrrhizin(GR) and boswellic acids (BA) in the treatment of COVID-19 but conclusive evidence is lacking. Our aim is to investigate the efficacy of GR + BA versus placebo for the treatment of hospitalized patients with moderate SARS-CoV-2 or COVID-19 variants infection. The current study is a randomized, double-blind, placebo-controlled, single-center trial. Patients with SARS-CoV-2 or COVID-19 variants diagnosed by PCR test who were admitted to Sohag University hospital were eligible if they were at least 18 years of age and had moderate symptoms. Patients were randomly assigned to receive oral GR capsule (60 mg) and BA (200 mg) twice daily for 14 days or a matching placebo. All patients also received treatment with the institutional protocol for COVID-19. The primary outcome was mortality and time to recovery. Secondary outcome was clinical status score, 14 days after receiving study drugs. Adverse events from use of study drugs have been evaluated for up to 14 days. The trial is registered at ClinicalTrials.gov (Identifier NCT04487964). During the 6-month enrollment period (June-November, 2021) only 50 patients (54% women; median age 60 years, IQR 54-65) met eligibility and were randomly assigned. Evaluation of the primary outcome at 14 days showed that there were five deaths in the placebo group and no deaths in the GR + BA group. With regard to recovery time, it was significantly shorter (p = 0.0001) in the group receiving GR + BA capsule compared to the placebo group (median 7.0; IQR 6.0-8.0 days vs. median 12.5; IQR 12-20 days). Clinical status on the ordinal score scale as a secondary outcome showed a significant difference between the GR + BA group (median (IQR) score, 2 [2-3]) and placebo groups (mean (IQR) score, 3 [3-5.5]). There was a significant decrease in CRB (p = 0.000041) in GR + BA compared with the placebo group. In conclusion, this safe, inexpensive, antiviral, immunomodulating and anti-inflammatory combination may be considered for use in mild to moderate infections of SARS-CoV-2 or COVID-19 variants. The study is limited by the small sample size; therefore, larger randomized trials are required.
Collapse
Affiliation(s)
- Adel A Gomaa
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Hamdy S Mohamed
- Department of Internal Medicine, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Rasha B Abd-Ellatief
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mohamed A Gomaa
- Department of Plastic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Doaa S Hammam
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
34
|
Darshani P, Sen Sarma S, Srivastava AK, Baishya R, Kumar D. Anti-viral triterpenes: a review. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2022; 21:1761-1842. [PMID: 35283698 PMCID: PMC8896976 DOI: 10.1007/s11101-022-09808-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/27/2022] [Indexed: 05/07/2023]
Abstract
Triterpenes are naturally occurring derivatives biosynthesized following the isoprene rule of Ruzicka. The triterpenes have been reported to possess a wide range of therapeutic applications including anti-viral properties. In this review, the recent studies (2010-2020) concerning the anti-viral activities of triterpenes have been summarized. The structure activity relationship studies have been described as well as brief biosynthesis of these triterpenes is discussed.
Collapse
Affiliation(s)
- Priya Darshani
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata, India
| | - Shreya Sen Sarma
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata, India
| | - Amit K. Srivastava
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata, India
| | - Rinku Baishya
- Natural Product Chemistry Group, CSIR-North East Institute of Science and Technology (NEIST), NH-37, Pulibor, Jorhat, Assam India
| | - Deepak Kumar
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata, India
| |
Collapse
|
35
|
Cho ES, Krishnan P, Loh HS, Daly JM, Leong CO, Mai CW, Low YY, Yong KT, Lim KH. Pentacyclic and hexacyclic cucurbitacins from Elaeocarpuspetiolatus. PHYTOCHEMISTRY 2022; 193:112988. [PMID: 34717280 DOI: 10.1016/j.phytochem.2021.112988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 02/08/2023]
Abstract
Four undescribed cucurbitacins, designated as petiolaticins A-D, and four known cucurbitacins were isolated from the bark and leaves of Elaeocarpus petiolatus (Jack) Wall. Their chemical structures were elucidated based on detailed analyses of the NMR and MS data. The absolute configuration of petiolaticin A was also determined by X-ray diffraction analysis. Petiolaticin A represents a cucurbitacin derivative incorporating a 3,4-epoxyfuranyl-bearing side chain, while petiolaticin B possesses a furopyranyl unit fused to the tetracyclic cucurbitane core structure. Petiolaticins A, B, and D were evaluated in vitro against a panel of human breast, pancreatic, and colorectal cancer cell lines. Petiolaticin A exhibited the greatest cytotoxicity against the MDA-MB-468, MDA-MB-231, MCF-7, and SW48 cell lines (IC50 7.4, 9.2, 9.3, and 4.6 μM, respectively). Additionally, petiolaticin D, 16α,23α-epoxy-3β,20β-dihydroxy-10αH,23βH-cucurbit-5,24-dien-11-one, and 16α,23α-epoxy-3β,20β-dihydroxy-10αH,23βH-cucurbit-5,24-dien-11-one 3-O-β-D-glucopyranoside were tested for their ability to inhibit cell entry of a pseudotyped virus bearing the hemagglutinin envelope protein of a highly pathogenic avian influenza virus. Petiolaticin D showed the highest inhibition (44.3%), followed by 16α,23α-epoxy-3β,20β-dihydroxy-10αH,23βH-cucurbit-5,24-dien-11-one (21.0%), and 16α,23α-epoxy-3β,20β-dihydroxy-10αH,23βH-cucurbit-5,24-dien-11-one 3-O-β-D-glucopyranoside showed limited inhibition (9.0%). These preliminary biological assays have demonstrated that petiolaticins A and D possess anticancer and antiviral properties, respectively, which warrant for further investigations.
Collapse
Affiliation(s)
- Eun-Seon Cho
- School of Biosciences, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Premanand Krishnan
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Hwei-San Loh
- School of Biosciences, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Janet M Daly
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, 57000 Kuala Lumpur, Malaysia; Centre for Cancer and Stem Cells Research, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cells Research, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia; State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yun-Yee Low
- Department of Chemistry, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kien-Thai Yong
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kuan-Hon Lim
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
36
|
Su S, Xu W, Jiang S. Virus Entry Inhibitors: Past, Present, and Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:1-13. [DOI: 10.1007/978-981-16-8702-0_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
37
|
Zhu J, Wang J, Wang X, Gao M, Guo B, Gao M, Liu J, Yu Y, Wang L, Kong W, An Y, Liu Z, Sun X, Huang Z, Zhou H, Zhang N, Zheng R, Xie Z. Prediction of drug efficacy from transcriptional profiles with deep learning. Nat Biotechnol 2021; 39:1444-1452. [PMID: 34140681 DOI: 10.1038/s41587-021-00946-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 05/06/2021] [Indexed: 02/05/2023]
Abstract
Drug discovery focused on target proteins has been a successful strategy, but many diseases and biological processes lack obvious targets to enable such approaches. Here, to overcome this challenge, we describe a deep learning-based efficacy prediction system (DLEPS) that identifies drug candidates using a change in the gene expression profile in the diseased state as input. DLEPS was trained using chemically induced changes in transcriptional profiles from the L1000 project. We found that the changes in transcriptional profiles for previously unexamined molecules were predicted with a Pearson correlation coefficient of 0.74. We examined three disorders and experimentally tested the top drug candidates in mouse disease models. Validation showed that perillen, chikusetsusaponin IV and trametinib confer disease-relevant impacts against obesity, hyperuricemia and nonalcoholic steatohepatitis, respectively. DLEPS can generate insights into pathogenic mechanisms, and we demonstrate that the MEK-ERK signaling pathway is a target for developing agents against nonalcoholic steatohepatitis. Our findings suggest that DLEPS is an effective tool for drug repurposing and discovery.
Collapse
Affiliation(s)
- Jie Zhu
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Jingxiang Wang
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China
| | - Xin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Mingjing Gao
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China
| | - Bingbing Guo
- Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking University, Beijing, China
| | - Miaomiao Gao
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking University, Beijing, China
| | - Yanqiu Yu
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China
| | - Liang Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Weikaixin Kong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Health Science Center, Peking University, Beijing, China
| | - Yongpan An
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Zurui Liu
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Health Science Center, Peking University, Beijing, China
| | - Hong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.
| | - Ning Zhang
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China.
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking University, Beijing, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China. .,Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China.
| |
Collapse
|
38
|
Kazakova O, Tret'yakova E, Baev D. Evaluation of A-azepano-triterpenoids and related derivatives as antimicrobial and antiviral agents. J Antibiot (Tokyo) 2021; 74:559-573. [PMID: 34253887 PMCID: PMC8273037 DOI: 10.1038/s41429-021-00448-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
A series of semisynthetic triterpenoids with A-ring azepano- and A-seco-fragments as well as hydrazido/hydrazono-substituents at C3 and C28 has been synthesized and evaluated for antimicrobial activity against key ESKAPE pathogens and DNA viruses (HSV-1, HCMV, HPV-11). It was found that azepanouvaol 8, 3-amino-3,4-seco-4(23)-en derivatives of uvaol 21 and glycyrrhetol-dien 22 as well as azepano-glycyrrhetol-tosylate 32 showed strong antimicrobial activities against MRSA with MIC ≤ 0.15 μM that exceeds the effect of antibiotic vancomycin. Azepanobetulinic acid cyclohexyl amide 4 exhibited significant bacteriostatic effect against MRSA (MIC ≤ 0.15 μM) with low cytotoxicity to HEK-293 even at a maximum tested concentration of >20 μM (selectivity index SI 133) and may be considered a noncytotoxic anti-MRSA agent. Azepanobetulin 1, azepanouvaol 8, and azepano-glycyrrhetol 15 showed high potency towards HCMV (EC50 0.15; 0.11; 0.11 µM) with selectivity indexes SI50 115; 136; 172, respectively. The docking studies suggest the possible interactions of the leading compounds with the molecular targets.
Collapse
Affiliation(s)
- Oxana Kazakova
- Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, 71 Prospect Oktyabrya, 450054, Ufa, Russian Federation.
| | - Elena Tret'yakova
- Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, 71 Prospect Oktyabrya, 450054, Ufa, Russian Federation
| | - Dmitry Baev
- N.N. Vorozhtzov Novosibirsk Institute of Organic Chemistry SB RAS, 9, Lavrent'ev Ave., Novosibirsk, 630090, Russian Federation
| |
Collapse
|
39
|
Li W, Ma X, Li G, Zhang A, Wang D, Fan F, Ma X, Zhang X, Dai Z, Qian Z. De Novo Biosynthesis of the Oleanane-Type Triterpenoids of Tunicosaponins in Yeast. ACS Synth Biol 2021; 10:1874-1881. [PMID: 34259519 DOI: 10.1021/acssynbio.1c00065] [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] [Indexed: 01/19/2023]
Abstract
Tunicosaponins are natural products extracted from Psammosilene tunicoides, which is an important ingredient of Yunnan Baiyao Powder, an ancient and famous Asian herbal medicine. The representative aglycones of tunicosaponins are the oleanane-type triterpenoids of gypsogenin and quillaic acid, which were found to manipulate a broad range of virus-host fusion via wrapping the heptad repeat-2 (HR2) domain prevalent in viral envelopes. However, the unknown biosynthetic pathway and difficulty in chemical synthesis hinder the therapeutic use of tunicosaponins. Here, two novel cytochrome P450-dependent monooxygenases that take part in the biosynthesis of tunicosaponins, CYP716A262 (CYP091) and CYP72A567 (CYP099), were identified from P. tunicoides. In addition, the whole biosynthesis pathway of the tunicosaponin aglycones was reconstituted in yeast by transforming the platform strain BY-bAS with the CYP716A262 and CYP716A567 genes, the resulting strain could produce 146.84 and 314.01 mg/L of gypsogenin and quillaic acid, respectively. This synthetic biology platform for complicated metabolic pathways elucidation and microbial cell factories construction can provide alternative sources of important natural products, helping conserve natural plant resources.
Collapse
Affiliation(s)
- Weixian Li
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming, 650500, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Department of Pharmacy, The Third People’s Hospital of Kunming, Kunming, 650000, China
| | - Xiaohui Ma
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Guodong Li
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Aili Zhang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Dong Wang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Feiyu Fan
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Xiaolin Ma
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Xueli Zhang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Zhubo Dai
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Zigang Qian
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming, 650500, China
| |
Collapse
|
40
|
Si L, Bai H, Rodas M, Cao W, Oh CY, Jiang A, Moller R, Hoagland D, Oishi K, Horiuchi S, Uhl S, Blanco-Melo D, Albrecht RA, Liu WC, Jordan T, Nilsson-Payant BE, Golynker I, Frere J, Logue J, Haupt R, McGrath M, Weston S, Zhang T, Plebani R, Soong M, Nurani A, Kim SM, Zhu DY, Benam KH, Goyal G, Gilpin SE, Prantil-Baun R, Gygi SP, Powers RK, Carlson KE, Frieman M, tenOever BR, Ingber DE. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng 2021; 5:815-829. [PMID: 33941899 PMCID: PMC8387338 DOI: 10.1038/s41551-021-00718-9] [Citation(s) in RCA: 254] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/19/2021] [Indexed: 02/05/2023]
Abstract
The rapid repurposing of antivirals is particularly pressing during pandemics. However, rapid assays for assessing candidate drugs typically involve in vitro screens and cell lines that do not recapitulate human physiology at the tissue and organ levels. Here we show that a microfluidic bronchial-airway-on-a-chip lined by highly differentiated human bronchial-airway epithelium and pulmonary endothelium can model viral infection, strain-dependent virulence, cytokine production and the recruitment of circulating immune cells. In airway chips infected with influenza A, the co-administration of nafamostat with oseltamivir doubled the treatment-time window for oseltamivir. In chips infected with pseudotyped severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), clinically relevant doses of the antimalarial drug amodiaquine inhibited infection but clinical doses of hydroxychloroquine and other antiviral drugs that inhibit the entry of pseudotyped SARS-CoV-2 in cell lines under static conditions did not. We also show that amodiaquine showed substantial prophylactic and therapeutic activities in hamsters challenged with native SARS-CoV-2. The human airway-on-a-chip may accelerate the identification of therapeutics and prophylactics with repurposing potential.
Collapse
Affiliation(s)
- Longlong Si
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Melissa Rodas
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Wuji Cao
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Crystal Yuri Oh
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Amanda Jiang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rasmus Moller
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daisy Hoagland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kohei Oishi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shu Horiuchi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Skyler Uhl
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Blanco-Melo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wen-Chun Liu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tristan Jordan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ilona Golynker
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Justin Frere
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James Logue
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert Haupt
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marisa McGrath
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stuart Weston
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tian Zhang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Roberto Plebani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Center on Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Mercy Soong
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Atiq Nurani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Seong Min Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Danni Y Zhu
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kambez H Benam
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sarah E Gilpin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Rani K Powers
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kenneth E Carlson
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Benjamin R tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
41
|
Gomaa AA, Mohamed HS, Abd-Ellatief RB, Gomaa MA. Boswellic acids/Boswellia serrata extract as a potential COVID-19 therapeutic agent in the elderly. Inflammopharmacology 2021; 29:1033-1048. [PMID: 34224069 PMCID: PMC8256410 DOI: 10.1007/s10787-021-00841-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/20/2021] [Indexed: 12/14/2022]
Abstract
The most severe cases of COVID-19, and the highest rates of death, are among the elderly. There is an urgent need to search for an agent to treat the disease and control its progression. Boswellia serrata is traditionally used to treat chronic inflammatory diseases of the lung. This review aims to highlight currently published research that has shown evidence of potential therapeutic effects of boswellic acids (BA) and B. serrata extract against COVID-19 and associated conditions. We reviewed the published information up to March 2021. Studies were collected through a search of online electronic databases (academic libraries such as PubMed, Scopus, Web of Science, and Egyptian Knowledge Bank). Several recent studies reported that BAs and B. serrata extract are safe agents and have multiple beneficial activities in treating similar symptoms experienced by patients with COVID-19. Because of the low oral bioavailability and improvement of buccal/oral cavity hygiene, traditional use by chewing B. serrata gum may be more beneficial than oral use. It is the cheapest option for a lot of poorer people. The promising effect of B. serrata and BA can be attributed to its antioxidant, anti-inflammatory, immunomodulatory, cardioprotective, anti-platelet aggregation, antibacterial, antifungal, and broad antiviral activity. B. serrata and BA act by multiple mechanisms. The most common mechanism may be through direct interaction with IκB kinases and inhibiting nuclear factor-κB-regulated gene expression. However, the most recent mechanism proposed that BA not only inhibited the formation of classical 5-lipoxygenase products but also produced anti-inflammatory LOX-isoform-selective modulators. In conclusion a small to moderate dose B. serrata extract may be useful in the enhancing adaptive immune response in mild to moderate symptoms of COVID-19. However, large doses of BA may be beneficial in suppressing uncontrolled activation of the innate immune response. More clinical results are required to determine with certainty whether there is sufficient evidence of the benefits against COVID-19.
Collapse
Affiliation(s)
- Adel A Gomaa
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Hamdy S Mohamed
- Department of Internal Medicine, Faculty of Medicine, Sohage University, Sohâg, Egypt
| | | | - Mohamed A Gomaa
- Department of Plastic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
42
|
Gomaa AA, Abdel-Wadood YA. The potential of glycyrrhizin and licorice extract in combating COVID-19 and associated conditions. PHYTOMEDICINE PLUS : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 1:100043. [PMID: 35399823 PMCID: PMC7886629 DOI: 10.1016/j.phyplu.2021.100043] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 04/28/2023]
Abstract
BACKGROUND Several recent studies have stated that glycyrrhizin and licorice extract are present in most traditional Chinese medicine formulas used against SARS-CoV-2 in China. Significant data are showing that glycyrrhizin and licorice extract have multiple beneficial activities in combating most features of SARS-CoV-2. PURPOSE The aim of current review was to highlight recent progresses in research that showed the evidence of the potential use of glycyrrhizin and licorice extract against COVID-19. METHODOLOGY We have reviewed the information published from 1979 to October 2020. These studies demonstrated the effects , use and safety of glycyrrhizin and icorice extract against viral infections,bacterial infections, inflammatory disorders of lung ( in vitro and in vivo). These studies were collated through online electronic databases research (Academic libraries as PubMed, Scopus, Web of Science and Egyptian Knowledge Bank). RESULTS Pooled effect size of articles provides information about the rationale for using glycyrrhizin and licorice extract to treat COVID-19. Fifty studies demonstrate antiviral activity of glycyrrhizin and licorice extract. The most frequent mechanism of the antiviral activity is due to disrupting viral uptake into the host cells and disrupting the interaction between receptor- binding domain (RBD) of SARS-COV2 and ACE2 in recent articles. Fifty studies indicate that glycyrrhizin and licorice extract have significant antioxidant, anti-inflammatory and immunomodulatory effects. Twenty five studies provide evidence for the protective effect of glycyrrhizin and licorice extract against inflammation-induced acute lung injury and cardiovascular disorders. CONCLUSION The current study showed several evidence regarding the beneficial effects of glycyrrhizin and licorice extract in combating COVID-19. More randomized clinical trials are needed to obtain a precise conclusion.
Collapse
Key Words
- 18β-GA, 18β-glycyrrhetinic acid
- : ACE2, angiotensin-converting enzyme 2
- ALI, acute lung injury
- ARDS, acute Respiratory Distress Syndrome
- Acute lung injury protector
- COVID-19
- COVID-19, Coronavirus disease 2019
- COX-2, cyclooxygenase-2
- DCs, dendritic cells
- Gl, glycyrrhizin
- Glycyrrhizin and licorice extract;Antiviral and antimicrobial, Anti-inflammatory and antioxidant
- HBsAg, hepatitis B surface antigen
- HCV, hepatitis C virus
- HMGB1, high-mobility group box 1
- IL, interleukin
- Immunododulator
- MAPKs, mitogen-activated protein kinases
- MERS, Middle East respiratory syndrome
- MR, mineralocorticoid receptor
- MRSA, Methicillin-resistant Staphylococcus aureus
- NO, nitric oxide
- RBD, receptor-binding domain
- ROS, reactive oxygen species
- S, Spike
- SARS, severe acute respiratory syndrome
- TCM, traditional Chinese medicine
- TLR, toll-like receptor
- TMPRSS2, type 2 transmembrane serine protease
- TNF-α, tumor necrosis factor alpha
- h, hour
- iNOS, inducible nitric oxide synthase
- licorice extract, LE
Collapse
Affiliation(s)
- Adel A Gomaa
- Department of Medical Pharmacology, Faculty of Medicine, Assiut Universitya, Beni-Suif, Egypt
| | | |
Collapse
|
43
|
Pu J, Zhou JT, Liu P, Yu F, He X, Lu L, Jiang S. Viral Entry Inhibitors Targeting Six-Helical Bundle Core Against Highly Pathogenic Enveloped Viruses with Class I Fusion Proteins. Curr Med Chem 2021; 29:700-718. [PMID: 33992055 DOI: 10.2174/0929867328666210511015808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 11/22/2022]
Abstract
TypeⅠ enveloped viruses bind to cell receptors through surface glycoproteins to initiate infection or undergo receptor-mediated endocytosis. They also initiate membrane fusion in the acidic environment of endocytic compartments, releasing genetic material into the cell. In the process of membrane fusion, envelope protein exposes fusion peptide, followed by insertion into the cell membrane or endosomal membrane. Further conformational changes ensue in which the type 1 envelope protein forms a typical six-helix bundle structure, shortening the distance between viral and cell membranes so that fusion can occur. Entry inhibitors targeting viral envelope proteins, or host factors, are effective antiviral agents and have been widely studied. Some have been used clinically, such as T20 and Maraviroc for human immunodeficiency virus 1 (HIV-1) or Myrcludex B for hepatitis D virus (HDV). This review focuses on entry inhibitors that target the six-helical bundle core against highly pathogenic enveloped viruses with class I fusion proteins, including retroviruses, coronaviruses, influenza A viruses, paramyxoviruses, and filoviruses.
Collapse
Affiliation(s)
- Jing Pu
- Key Laboratory of Medical Molecular Virology of MOE/MOH/CAMS, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China
| | - Joey Tianyi Zhou
- Institute of High Performance Computing, The Agency for Science, Technology and Research, Singapore
| | - Ping Liu
- Institute of High Performance Computing, The Agency for Science, Technology and Research, Singapore
| | - Fei Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Xiaoyang He
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of MOE/MOH/CAMS, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH/CAMS, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
44
|
Lu L, Su S, Yang H, Jiang S. Antivirals with common targets against highly pathogenic viruses. Cell 2021; 184:1604-1620. [PMID: 33740455 DOI: 10.1016/j.cell.2021.02.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/15/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Historically, emerging viruses appear constantly and have cost millions of human lives. Currently, climate change and intense globalization have created favorable conditions for viral transmission. Therefore, effective antivirals, especially those targeting the conserved protein in multiple unrelated viruses, such as the compounds targeting RNA-dependent RNA polymerase, are urgently needed to combat more emerging and re-emerging viruses in the future. Here we reviewed the development of antivirals with common targets, including those against the same protein across viruses, or the same viral function, to provide clues for development of antivirals for future epidemics.
Collapse
Affiliation(s)
- Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Shan Su
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China.
| |
Collapse
|
45
|
Wang HQ, Ma SG, Zhang D, Li YH, Qu J, Li Y, Liu YB, Yu SS. Oxygenated pentacyclic triterpenoids from the stems and branches of Enkianthus chinensis. Bioorg Chem 2021; 111:104866. [PMID: 33866237 DOI: 10.1016/j.bioorg.2021.104866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 11/28/2022]
Abstract
Thirty new pentacyclic triterpenoids, including five oleanane-type (1-5), twenty-three ursane-type (9-23, 26-33) and two taraxerane-type (24 and 25), along with fourteen known triterpenoids, were isolated from the stems and branches of Enkianthus chinensis. Their structures were elucidated by extensive spectroscopic analyses, X-ray crystallographic data and electronic circular dichroism (ECD) techniques. Sixteen compounds (1-5, 9-13, 20, 22, 32, 34-36) bearing a gem-hydroxymethyl group at C-4 represent rare examples of pentacyclic triterpenoids. In the in vitro biological activity evaluation, compounds 8, 9, 12-14, 17, 24, and 44 exhibited potent hepatoprotective effects at 10 μM. Moreover, compound 25 showed latent activity against HSV-1 with an IC50 value of 6.4 μM.
Collapse
Affiliation(s)
- Hai-Qiang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuang-Gang Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yu-Huan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Jing Qu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yong Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yun-Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shi-Shan Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China.
| |
Collapse
|
46
|
Formulation, Stability, Pharmacokinetic, and Modeling Studies for Tests of Synergistic Combinations of Orally Available Approved Drugs against Ebola Virus In Vivo. Microorganisms 2021; 9:microorganisms9030566. [PMID: 33801811 PMCID: PMC7998926 DOI: 10.3390/microorganisms9030566] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/28/2022] Open
Abstract
Outbreaks of Ebola ebolavirus (EBOV) have been associated with high morbidity and mortality. Milestones have been reached recently in the management of EBOV disease (EVD) with licensure of an EBOV vaccine and two monoclonal antibody therapies. However, neither vaccines nor therapies are available for other disease-causing filoviruses. In preparation for such outbreaks, and for more facile and cost-effective management of EVD, we seek a cocktail containing orally available and room temperature stable drugs with strong activity against multiple filoviruses. We previously showed that (bepridil + sertraline) and (sertraline + toremifene) synergistically suppress EBOV in cell cultures. Here, we describe steps towards testing these combinations in a mouse model of EVD. We identified a vehicle suitable for oral delivery of the component drugs and determined that, thus formulated the drugs are equally active against EBOV as preparations in DMSO, and they maintain activity upon storage in solution for up to seven days. Pharmacokinetic (PK) studies indicated that the drugs in the oral delivery vehicle are well tolerated in mice at the highest doses tested. Collectively the data support advancement of these combinations to tests for synergy in a mouse model of EVD. Moreover, mathematical modeling based on human oral PK projects that the combinations would be more active in humans than their component single drugs.
Collapse
|
47
|
Li H, Cheng C, Li S, Wu Y, Liu Z, Liu M, Chen J, Zhong Q, Zhang X, Liu S, Song G. Discovery and structural optimization of 3-O-β-chacotriosyl oleanane-type triterpenoids as potent entry inhibitors of SARS-CoV-2 virus infections. Eur J Med Chem 2021; 215:113242. [PMID: 33588180 PMCID: PMC7869707 DOI: 10.1016/j.ejmech.2021.113242] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 12/15/2022]
Abstract
Currently, SARS-CoV-2 virus is an emerging pathogen that has posed a serious threat to public health worldwide. However, no agents have been approved to treat SARS-CoV-2 infections to date, underscoring the great need for effective and practical therapies for SARS-CoV-2 outbreaks. We reported that a focused screen of OA saponins identified 3-O-β-chacotriosyl OA benzyl ester 2 as a novel small molecule inhibitor of SARS-CoV-2 virus entry, via binding to SARS-CoV-2 glycoprotein (S). We performed structure-activity relationship profiling of 2 and discovered C-17-COOH of OA was an important modification site that improved both inhibitor potency toward SARS-CoV-2 and selectivity index. Then optimization from hit to lead resulted in a potent fusion inhibitor 12f displaying strong inhibition against infectious SARS-CoV-2 with an IC50 value of 0.97 μM in vitro. Mechanism studies confirmed that inhibition of SARS-CoV-2 viral entry of 12f was mediated by the direct interaction with SARS-CoV-2 S2 subunit to block membrane fusion. These 3-O-β-chacotriosyl OA amide saponins are suitable for further optimization as SARS-CoV-2 entry inhibitors with the potential to be developed as therapeutic agents for the treatment of SARS-CoV-2 virus infections.
Collapse
Affiliation(s)
- Hui Li
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Chen Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sumei Li
- Department of Human Anatomy, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafet Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhihao Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Mingjian Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qiuyu Zhong
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Xuesha Zhang
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, 510515, China.
| | - Gaopeng Song
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
48
|
Schafer A, Xiong R, Cooper L, Nowar R, Lee H, Li Y, Ramirez BE, Peet NP, Caffrey M, Thatcher GRJ, Saphire EO, Cheng H, Rong L. Evidence for distinct mechanisms of small molecule inhibitors of filovirus entry. PLoS Pathog 2021; 17:e1009312. [PMID: 33539432 PMCID: PMC7888603 DOI: 10.1371/journal.ppat.1009312] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/17/2021] [Accepted: 01/14/2021] [Indexed: 12/18/2022] Open
Abstract
Many small molecules have been identified as entry inhibitors of filoviruses. However, a lack of understanding of the mechanism of action for these molecules limits further their development as anti-filoviral agents. Here we provide evidence that toremifene and other small molecule entry inhibitors have at least three distinctive mechanisms of action and lay the groundwork for future development of anti-filoviral agents. The three mechanisms identified here include: (1) direct binding to the internal fusion loop region of Ebola virus glycoprotein (GP); (2) the HR2 domain is likely the main binding site for Marburg virus GP inhibitors and a secondary binding site for some EBOV GP inhibitors; (3) lysosome trapping of GP inhibitors increases drug exposure in the lysosome and further improves the viral inhibition. Importantly, small molecules targeting different domains on GP are synergistic in inhibiting EBOV entry suggesting these two mechanisms of action are distinct. Our findings provide important mechanistic insights into filovirus entry and rational drug design for future antiviral development.
Collapse
Affiliation(s)
- Adam Schafer
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Rui Xiong
- Department of Pharmaceutical Sciences, College of Pharmacy, and UICentre, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Laura Cooper
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America.,Department of Pharmaceutical Sciences, College of Pharmacy, and UICentre, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Raghad Nowar
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America.,Department of Pharmaceutical Sciences, College of Pharmacy, and UICentre, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Hyun Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, and UICentre, University of Illinois at Chicago, Chicago, Illinois, United States of America.,Biophysics core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Yangfeng Li
- Department of Pharmaceutical Sciences, College of Pharmacy, and UICentre, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Benjamin E Ramirez
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America.,NMR Core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Norton P Peet
- Chicago BioSolutions Inc., Chicago, Illinois, United States of America
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Gregory R J Thatcher
- Department of Pharmaceutical Sciences, College of Pharmacy, and UICentre, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | | | - Han Cheng
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
49
|
Christy MP, Uekusa Y, Gerwick L, Gerwick WH. Natural Products with Potential to Treat RNA Virus Pathogens Including SARS-CoV-2. JOURNAL OF NATURAL PRODUCTS 2021; 84:161-182. [PMID: 33352046 PMCID: PMC7771248 DOI: 10.1021/acs.jnatprod.0c00968] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Indexed: 05/03/2023]
Abstract
Three families of RNA viruses, the Coronaviridae, Flaviviridae, and Filoviridae, collectively have great potential to cause epidemic disease in human populations. The current SARS-CoV-2 (Coronaviridae) responsible for the COVID-19 pandemic underscores the lack of effective medications currently available to treat these classes of viral pathogens. Similarly, the Flaviviridae, which includes such viruses as Dengue, West Nile, and Zika, and the Filoviridae, with the Ebola-type viruses, as examples, all lack effective therapeutics. In this review, we present fundamental information concerning the biology of these three virus families, including their genomic makeup, mode of infection of human cells, and key proteins that may offer targeted therapies. Further, we present the natural products and their derivatives that have documented activities to these viral and host proteins, offering hope for future mechanism-based antiviral therapeutics. By arranging these potential protein targets and their natural product inhibitors by target type across these three families of virus, new insights are developed, and crossover treatment strategies are suggested. Hence, natural products, as is the case for other therapeutic areas, continue to be a promising source of structurally diverse new anti-RNA virus therapeutics.
Collapse
Affiliation(s)
- Mitchell P. Christy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - Yoshinori Uekusa
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Lena Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - William H. Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
50
|
Li H, Sun J, Xiao S, Zhang L, Zhou D. Triterpenoid-Mediated Inhibition of Virus-Host Interaction: Is Now the Time for Discovering Viral Entry/Release Inhibitors from Nature? J Med Chem 2020; 63:15371-15388. [PMID: 33201699 DOI: 10.1021/acs.jmedchem.0c01348] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fatal infectious diseases caused by HIV-1, influenza A virus, Ebola virus, and currently pandemic coronavirus highlight the great need for the discovery of antiviral agents in mechanisms different from current viral replication-targeted approaches. Given the critical role of virus-host interactions in the viral life cycle, the development of entry or shedding inhibitors may expand the current repertoire of antiviral agents; the combination of antireplication inhibitors and entry or shedding inhibitors would create a multifaceted drug cocktail with a tandem antiviral mechanism. Therefore, we provide critical information about triterpenoids as potential antiviral agents targeting entry and release, focusing specifically on the emerging aspect of triterpenoid-mediated inhibition of a variety of virus-host membrane fusion mechanisms via a trimer-of-hairpin motif. These properties of triterpenoids supply their host an evolutionary advantage for chemical defense and may protect against an increasingly diverse array of viruses infecting mammals, providing a direction for antiviral drug discovery.
Collapse
Affiliation(s)
- Haiwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing 100191, China
| | - Jiaqi Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing 100191, China
| | - Sulong Xiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing 100191, China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing 100191, China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing 100191, China
| |
Collapse
|