1
|
Khandoker Minu M, Enamul Kabir Talukder M, Mothana RA, Injamamul Islam S, Alanzi AR, Hasson S, Irfan Sadique M, Arfat Raihan Chowdhury M, Shajid Khan M, Ahammad F, Mohammad F. In-vitro and in-silico evaluation of rue herb for SARS-CoV-2 treatment. Int Immunopharmacol 2024; 143:113318. [PMID: 39393270 DOI: 10.1016/j.intimp.2024.113318] [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: 06/02/2024] [Revised: 08/27/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024]
Abstract
SARS-CoV-2, a β-coronavirus responsible for the COVID-19 pandemic, has resulted in approximately 4.9 million fatalities worldwide. Despite the urgent need, there is currently no specific therapeutic developed for treating or preventing SARS-CoV-2 infections. The virus enters the host by engaging in a molecular interaction between the viral Spike glycoprotein (S protein) and the host ACE2 receptor, facilitating membrane fusion and initiating infection. Inhibiting this interaction could impede viral activity. Therefore, this study aimed to identify natural small molecules from perennial rue herb (Ruta graveolens) as potential inhibitors against the S protein, thus preventing virus infection. Initially, a screening process was conducted on 53 compounds identified from rue herbs, utilizing pharmacophore-based virtual screening approaches. This analysis resulted in the identification of 12 hit compounds. Four compounds, namely Amentoflavone (CID: 5281600), Agathisflavone (CID: 5281599), Vitamin P (CID: 24832108), and Daphnoretin (CID: 5281406), emerged as potential S protein inhibitors through molecular docking simulations, exhibiting binding energies in kcal/mol of -9.2, -8.8, -8.2, and -8.0, respectively. ADMET analysis revealed favorable pharmacokinetics and toxicity profiles for these compounds. The compounds' stability with respect to the target S protein was evaluated using MD simulation and MM-GBSA approaches. The analysis revealed the stability of the selected compounds with the target protein. Also, PCA revealed distinctive movement patterns in four selected compounds, offered valuable insights into their functional behaviors and potential interactions. In-vitro assays revealed that rue herb extracts containing these compounds displayed potential inhibitory properties against the virus, with an IC50 value of 1.299 mg/mL and a cytotoxic concentration (CC50) value of 11.991 mg/mL. The compounds derived from rue herb, specifically Amentoflavone, Agathisflavone, Vitamin P, and Daphnoretin, show promise as candidates for the therapeutic intervention of SARS-CoV-2-related complications.
Collapse
Affiliation(s)
- Maliha Khandoker Minu
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7430, Bangladesh
| | - Md Enamul Kabir Talukder
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7430, Bangladesh
| | - Ramzi A Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sk Injamamul Islam
- The International Graduate Program of Veterinary Science and Technology (VST), Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Abdullah R Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sidgi Hasson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L33AF, UK
| | - Md Irfan Sadique
- Department of Biological Science, Carnegie Mellon University 24866 Doha, Qatar
| | | | - Md Shajid Khan
- Chemical Engineering Program, Texas A&M University at Qatar, Doha 4290, Qatar
| | - Foysal Ahammad
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7430, Bangladesh; Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha 34110, Qatar.
| | - Farhan Mohammad
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha 34110, Qatar.
| |
Collapse
|
2
|
Tenginakai P, Bhor S, Waasia FZ, Sharma S, Dinesh S. Comparative proteomic analysis to annotate the structural association of the hypothetical proteins from the conserved domain of P. aeruginosa as novel vaccine candidates. Biotechnol Lett 2024; 47:13. [PMID: 39702823 DOI: 10.1007/s10529-024-03546-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/18/2024] [Accepted: 10/03/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVES Pseudomonas aeruginosa, identified as an ESKAPE pathogen, contributes to severe clinical diseases worldwide and despite its prevalence an effective vaccine or treatment remains elusive. Numerous computational methods are being employed to target hypothetical proteins (HPs). Presently, no studies have predicted multi-epitope vaccines for these HPs. RESULTS Totally, 877 HPs from P. aeruginosa were included in the study and the data showcased here illustrate a methodical approach to prioritize the proteome by employing diverse comparative proteomics. The study employed physicochemical property assessment and conserved domain analysis to identify stable and immunologically pertinent proteins for epitope prediction. The VaxiJen2.0 antigenicity assessment aided in epitope selection, contributing to the foundational steps in vaccine development by predicting T-cell and B-cell epitopes. Potential T and B cell epitopes with high antigenicity, non-toxic categorization, and robust binding affinities were identified in the investigation. The periplasmic HP WP_132813935.1 was predicted as conserved, stable, and soluble. The T-cell peptide RTSMRALAY and the B-cell peptide MPVYLYLM were predicted to be probable non-allergen and demonstrated strong binding with MHC class I allele HLA-C*03:03. CONCLUSIONS This research provides a comprehensive approach to predict T and B cell epitopes for conditions associated with P. aeruginosa, offering a candidate pool for tailored vaccine development. However, the efficacy of these epitopes in vaccine development necessitates clinical validation and testing for confirmation.
Collapse
Affiliation(s)
- Prajval Tenginakai
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India
| | - Samiksha Bhor
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India
| | | | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India.
| |
Collapse
|
3
|
Asadi M, Ghasemi Y, Nezafat N, Sarkari B, Baneshi M, Mostafavi-Pour Z, Anbardar MH, Savardashtaki A. Designing a novel multi-epitope antigen for diagnosing human cytomegalovirus infection: An immunoinformatics approach. Biotechnol Appl Biochem 2024. [PMID: 39417400 DOI: 10.1002/bab.2677] [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: 01/25/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
Human cytomegalovirus (HCMV) infection can lead to congenital infections and severe complications, particularly in immunocompromised individuals. Current serological tests for diagnosing HCMV infection often face limitations in sensitivity and specificity. Developing multi-epitope antigens for serological assays offers the potential for enhancing diagnostic accuracy. This study aimed to design a novel multi-epitope antigen for HCMV infection diagnosis using immunoinformatic approaches. Five tegument proteins (universal protein resource [UniProt] ID: Po8318, Po6725, F5HC97, Q6RX10, and F5HC05) were selected based on their antigenic properties and literature review. Six linear B-cell epitopes were predicted within conserved regions of each antigen sequence and linked with appropriate linkers. The designed multi-epitope antigen underwent thorough evaluation for physicochemical properties, solubility, antigenicity, and cross-reactivity. Additionally, the three-dimensional structure of the antigen was predicted, refined, and validated. The nucleotide sequence of the designed antigen was optimized for successful expression in Escherichia coli and inserted into a pET23a (+) vector. Immunoinformatic analysis revealed that the multi-epitope antigen exhibits stability, antigenicity, and lacks cross-reactivity. Our findings suggest that this multi-epitope antigen is a promising candidate for diagnosing HCMV infection. However, further validation through laboratory testing is required to confirm its diagnostic efficacy.
Collapse
Affiliation(s)
- Marzieh Asadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Navid Nezafat
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Bahador Sarkari
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Maryam Baneshi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Shiraz, Iran
| | - Zohreh Mostafavi-Pour
- Recombinant Protein Laboratory, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Hossain MR, Tareq MMI, Biswas P, Tauhida SJ, Bibi S, Zilani MNH, Albadrani GM, Al‐Ghadi MQ, Abdel‐Daim MM, Hasan MN. Identification of molecular targets and small drug candidates for Huntington's disease via bioinformatics and a network-based screening approach. J Cell Mol Med 2024; 28:e18588. [PMID: 39153206 PMCID: PMC11330274 DOI: 10.1111/jcmm.18588] [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: 05/18/2024] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
Huntington's disease (HD) is a gradually severe neurodegenerative ailment characterised by an increase of a specific trinucleotide repeat sequence (cytosine-adenine-guanine, CAG). It is passed down as a dominant characteristic that worsens over time, creating a significant risk. Despite being monogenetic, the underlying mechanisms as well as biomarkers remain poorly understood. Furthermore, early detection of HD is challenging, and the available diagnostic procedures have low precision and accuracy. The research was conducted to provide knowledge of the biomarkers, pathways and therapeutic targets involved in the molecular processes of HD using informatic based analysis and applying network-based systems biology approaches. The gene expression profile datasets GSE97100 and GSE74201 relevant to HD were studied. As a consequence, 46 differentially expressed genes (DEGs) were identified. 10 hub genes (TPM1, EIF2S3, CCN2, ACTN1, ACTG2, CCN1, CSRP1, EIF1AX, BEX2 and TCEAL5) were further differentiated in the protein-protein interaction (PPI) network. These hub genes were typically down-regulated. Additionally, DEGs-transcription factors (TFs) connections (e.g. GATA2, YY1 and FOXC1), DEG-microRNA (miRNA) interactions (e.g. hsa-miR-124-3p and has-miR-26b-5p) were also comprehensively forecast. Additionally, related gene ontology concepts (e.g. sequence-specific DNA binding and TF activity) connected to DEGs in HD were identified using gene set enrichment analysis (GSEA). Finally, in silico drug design was employed to find candidate drugs for the treatment HD, and while the possible modest therapeutic compounds (e.g. cortistatin A, 13,16-Epoxy-25-hydroxy-17-cheilanthen-19,25-olide, Hecogenin) against HD were expected. Consequently, the results from this study may give researchers useful resources for the experimental validation of Huntington's diagnosis and therapeutic approaches.
Collapse
Affiliation(s)
- Md Ridoy Hossain
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Md. Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Sadia Jannat Tauhida
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| | - Shabana Bibi
- Department of BiosciencesShifa Tameer‐e‐Millat UniversityIslamabadPakistan
- Department of Health SciencesNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | | | - Ghadeer M. Albadrani
- Department of Biology, College of SciencePrincess Nourah bint Abdulrahman UniversityRiyadhSaudi Arabia
| | - Muath Q. Al‐Ghadi
- Department of Zoology, College of ScienceKing Saud UniversityRiyadhSaudi Arabia
| | - Mohamed M. Abdel‐Daim
- Department of Pharmaceutical Sciences, Pharmacy ProgramBatterjee Medical CollegeJeddahSaudi Arabia
- Pharmacology Department, Faculty of Veterinary MedicineSuez Canal UniversityIsmailiaEgypt
| | - Md. Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJessoreBangladesh
| |
Collapse
|
5
|
Kolla HB, Dutt M, Kumar A, Hebbandi Nanjunadappa R, Karakach T, Singh KP, Kelvin D, Clement Mertens PP, Umeshappa CS. Immuno-informatics study identifies conserved T cell epitopes in non-structural proteins of Bluetongue virus serotypes: formulation of a computationally optimized next-generation broad-spectrum multi-epitope vaccine. Front Immunol 2024; 15:1424307. [PMID: 39011043 PMCID: PMC11246920 DOI: 10.3389/fimmu.2024.1424307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction Bluetongue (BT) poses a significant threat to the livestock industry, affecting various animal species and resulting in substantial economic losses. The existence of numerous BT virus (BTV) serotypes has hindered control efforts, highlighting the need for broad-spectrum vaccines. Methodology In this study, we evaluated the conserved amino acid sequences within key non-structural (NS) proteins of BTV and identified numerous highly conserved murine- and bovine-specific MHC class I-restricted (MHC-I) CD8+ and MHC-II-restricted CD4+ epitopes. We then screened these conserved epitopes for antigenicity, allergenicity, toxicity, and solubility. Using these epitopes, we developed in silico-based broad-spectrum multiepitope vaccines with Toll-like receptor (TLR-4) agonists. The predicted proinflammatory cytokine response was assessed in silico using the C-IMMSIM server. Structural modeling and refinement were achieved using Robetta and GalaxyWEB servers. Finally, we assessed the stability of the docking complexes through extensive 100-nanosecond molecular dynamics simulations before considering the vaccines for codon optimization and in silico cloning. Results We found many epitopes that meet these criteria within NS1 and NS2 proteins and developed in silico broad-spectrum vaccines. The immune simulation studies revealed that these vaccines induce high levels of IFN-γ and IL-2 in the vaccinated groups. Protein-protein docking analysis demonstrated promising epitopes with strong binding affinities to TLR-4. The docked complexes were stable, with minimal Root Mean Square Deviation and Root Mean Square Fluctuation values. Finally, the in silico-cloned plasmids have high % of GC content with > 0.8 codon adaptation index, suggesting they are suitable for expressing the protein vaccines in prokaryotic system. Discussion These next-generation vaccine designs are promising and warrant further investigation in wet lab experiments to assess their immunogenicity, safety, and efficacy for practical application in livestock. Our findings offer a robust framework for developing a comprehensive, broad-spectrum vaccine, potentially revolutionizing BT control and prevention strategies in the livestock industry.
Collapse
Affiliation(s)
- Harish Babu Kolla
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Mansi Dutt
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Anuj Kumar
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Roopa Hebbandi Nanjunadappa
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Tobias Karakach
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Karam Pal Singh
- Center for Animal Disease Research and Diagnosis, Indian Veterinary Research Institute, Bareilly, India
| | - David Kelvin
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | | | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| |
Collapse
|
6
|
Gambichler T, Schrama D, Käpynen R, Weyer-Fahlbusch SS, Becker JC, Susok L, Kreppel F, Abu Rached N. Current Progress in Vaccines against Merkel Cell Carcinoma: A Narrative Review and Update. Vaccines (Basel) 2024; 12:533. [PMID: 38793784 PMCID: PMC11125734 DOI: 10.3390/vaccines12050533] [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: 03/19/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Merkel cell carcinoma is a rare, aggressive skin cancer that mainly occurs in elderly and immunocompromised patients. Due to the success of immune checkpoint inhibition in MCC, the importance of immunotherapy and vaccines in MCC has increased in recent years. In this article, we aim to present the current progress and perspectives in the development of vaccines for this disease. Here, we summarize and discuss the current literature and ongoing clinical trials investigating vaccines against MCC. We identified 10 articles through a PubMed search investigating a vaccine against MCC. From the international clinical trial database Clinical.Trials.gov, we identified nine studies on vaccines for the management of MCC, of which seven are actively recruiting. Most of the identified studies investigating a vaccine against MCC are preclinical or phase 1/2 trials. The vaccine principles mainly included DNA- and (synthetic) peptide-based vaccines, but RNA-based vaccines, oncolytic viruses, and the combination of vaccines and immunotherapy are also under investigation for the treatment of MCC. Although the management of MCC is changing, when compared to times before the approval of immune checkpoint inhibitors, it will still take some time before the first MCC vaccine is ready for approval.
Collapse
Affiliation(s)
- Thilo Gambichler
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany; (R.K.); (N.A.R.)
- Department of Dermatology, Dortmund Hospital gGmbH and Faculty of Health, Witten-Herdecke University, 44122 Dortmund, Germany; (S.S.W.-F.); (L.S.)
- Department of Dermatology and Phlebology, Christian Hospital Unna, 59423 Unna, Germany
| | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Riina Käpynen
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany; (R.K.); (N.A.R.)
| | - Sera S. Weyer-Fahlbusch
- Department of Dermatology, Dortmund Hospital gGmbH and Faculty of Health, Witten-Herdecke University, 44122 Dortmund, Germany; (S.S.W.-F.); (L.S.)
| | - Jürgen C. Becker
- Translational Skin Cancer Research, DKTK Partner Site Essen/Düsseldorf, West German Cancer Center, Department of Dermatology, University Duisburg-Essen, 45122 Essen, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Laura Susok
- Department of Dermatology, Dortmund Hospital gGmbH and Faculty of Health, Witten-Herdecke University, 44122 Dortmund, Germany; (S.S.W.-F.); (L.S.)
| | - Florian Kreppel
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany;
| | - Nessr Abu Rached
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany; (R.K.); (N.A.R.)
| |
Collapse
|
7
|
Munna MMR, Islam MA, Shanta SS, Monty MA. Structural, functional, molecular docking analysis of a hypothetical protein from Talaromyces marneffei and its molecular dynamic simulation: an in-silico approach. J Biomol Struct Dyn 2024:1-20. [PMID: 38345137 DOI: 10.1080/07391102.2024.2314264] [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: 07/31/2023] [Accepted: 01/29/2024] [Indexed: 03/01/2024]
Abstract
Telaromyces marneffei (formerly Penicillium marneffei) is an endemic pathogenic fungus in Southern China and Southeast Asia. It can cause disease in patients with travel-related exposure to this organism and high morbidity and mortality in acquired immune deficiency syndrome (AIDS). In this study, we analyzed the structure and function of a hypothetical protein from T. marneffei using several bioinformatics tools and servers to unveil novel pharmacological targets and design a peptide vaccine against specific epitopes. A total of seven functional epitopes were screened on the protein, and 'STGVDMWSV' was the most antigenic, non-allergenic and non-toxic. Molecular docking showed stronger affinity between the CTL epitope 'STGVDMWSV' and the MHC I allele HLA-A*02:01, a higher docking score -234.98 kcal/mol, revealed stable interactions during a 100 ns molecular dynamic simulation. Overall, the results of this study revealed that this hypothetical protein is crucial for comprehending biochemical, physiological pathways and identifying novel therapeutic targets for human health. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Md Masudur Rahman Munna
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Ariful Islam
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Saima Sajnin Shanta
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Masuma Akter Monty
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, PR China
| |
Collapse
|