1
|
Leal ES, Pascual MJ, Adler NS, Arrupe N, Merwaiss F, Giordano L, Fidalgo D, Álvarez D, Bollini M. Unveiling tetrahydroquinolines as promising BVDV entry inhibitors: Targeting the envelope protein. Virology 2024; 590:109968. [PMID: 38141499 DOI: 10.1016/j.virol.2023.109968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/25/2023]
Abstract
Bovine viral diarrhea virus (BVDV) is known to cause financial losses and decreased productivity in the cattle industry worldwide. Currently, there are no available antiviral treatments for effectively controlling BVDV infections in laboratories or farms. The BVDV envelope protein (E2) mediates receptor recognition on the cell surface and is required for fusion of virus and cell membranes after the endocytic uptake of the virus during the entry process. Therefore, E2 is an attractive target for the development of antiviral strategies. To identify BVDV antivirals targeting E2 function, we defined a binding site in silico located in domain IIIc at the interface between monomers in the disulfide linked dimer of E2. Employing a de novo design methodology to identify compounds with the potential to inhibit the E2 function, compound 9 emerged as a promising candidate with remarkable antiviral activity and minimal toxicity. In line with targeting of E2 function, compound 9 was found to block the virus entry into host cells. Furthermore, we demonstrated that compound 9 selectively binds to recombinant E2 in vitro. Molecular dynamics simulations (MD) allowed describing a possible interaction pattern between compound 9 and E2 and indicated that the S enantiomer of compound 9 may be responsible for the antiviral activity. Future research endeavors will focus on synthesizing enantiomerically pure compounds to further support these findings. These results highlight the usefulness of de novo design strategies to identify a novel class of BVDV inhibitors that block E2 function inhibiting virus entry into the host cell.
Collapse
Affiliation(s)
- Emilse S Leal
- Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María J Pascual
- Instituto de Investigaciones Biotecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Martín, Argentina
| | - Natalia S Adler
- Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Nicolás Arrupe
- Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Fernando Merwaiss
- Instituto de Investigaciones Biotecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Martín, Argentina
| | - Luciana Giordano
- Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniela Fidalgo
- Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Diego Álvarez
- Instituto de Investigaciones Biotecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Martín, Argentina.
| | - Mariela Bollini
- Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
2
|
Yang G, Wang J, Wang S, Zhu Y. Forsythiaside A Improves the Inhibitory Efficiency of Recombinant Protein Vaccines against Bovine Viral Diarrhea Virus Infection. Int J Mol Sci 2022; 23:ijms23169390. [PMID: 36012654 PMCID: PMC9409473 DOI: 10.3390/ijms23169390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) is a critical animal pathogen that leads to cattle production losses associated with acute disease, immune dysregulation, reproductive failure, and respiratory disease. Due to the monotonous control technique and neglect of BVDV, increasing prevalence of BVDV has caused significant economic losses in the cattle industry worldwide. Therefore, novel anti-BVDV drugs are essential to prevent and control BVDV. Our previous studies have found that Forsythoside A (FTA) could inhibit the replication of BVDV via TRAF2-dependent CD28-4-1BB signaling in bovine peripheral blood mononuclear cells (PBMCs), but whether they can directly inhibit the BVDV remains unclear. Here, we further investigated the effects of FTA on BVDV and its underlying mechanisms of action. We found that FTA significantly inhibited the replication of BVDV in the MDBK cell directly. The results demonstrated that FTA could reduce the functional activation of Caspase-1 to inhibit the inflammatory response caused by BVDV infection and increase the expression of type I interferon (IFN-I) to clear the virus in vitro. The animal experiment was performed to evaluate the antiviral effect of FTA in vivo. Notably, after challenged with BVDV, mice with FTA + Erns-E2 protein displayed alleviated pathological damage and decreased the viral load in the spleen compared with mice inoculated with Erns-E2 protein. Furthermore, treatment with FTA enhanced body defense and delayed infection by the BVDV. Our results reveal that FTA suppresses BVDV replication both in vitro and in vivo and therefore shows promise as an anti-BVDV agent.
Collapse
Affiliation(s)
- Guanghui Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiufeng Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shenghua Wang
- OIE Porcine Reproductive and Respiratory Syndrome Reference Laboratory, China Animal Disease Control Center, Beijing 102629, China
- Correspondence: (S.W.); (Y.Z.)
| | - Yaohong Zhu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Correspondence: (S.W.); (Y.Z.)
| |
Collapse
|
3
|
Ibba R, Riu F, Delogu I, Lupinu I, Carboni G, Loddo R, Piras S, Carta A. Benzimidazole-2-Phenyl-Carboxamides as Dual-Target Inhibitors of BVDV Entry and Replication. Viruses 2022; 14:v14061300. [PMID: 35746771 PMCID: PMC9231222 DOI: 10.3390/v14061300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 11/26/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV), also known as Pestivirus A, causes severe infection mostly in cattle, but also in pigs, sheep and goats, causing huge economical losses on agricultural farms every year. The infections are actually controlled by isolation of persistently infected animals and vaccination, but no antivirals are currently available to control the spread of BVDV on farms. BVDV binds the host cell using envelope protein E2, which has only recently been targeted in the research of a potent and efficient antiviral. In contrast, RdRp has been successfully inhibited by several classes of compounds in the last few decades. As a part of an enduring antiviral research agenda, we designed a new series of derivatives that emerged from an isosteric substitution of the main scaffold in previously reported anti-BVDV compounds. Here, the new compounds were characterized and tested, where several turned out to be potent and selectively active against BVDV. The mechanism of action was thoroughly studied using a time-of-drug-addition assay and the results were validated using docking simulations.
Collapse
Affiliation(s)
- Roberta Ibba
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (R.I.); (F.R.); (I.L.); (A.C.)
| | - Federico Riu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (R.I.); (F.R.); (I.L.); (A.C.)
| | - Ilenia Delogu
- Department of Biomedical Sciences, Cittadella Universitaria Monserrato, University of Cagliari, 09042 Monserrato, Italy;
| | - Ilenia Lupinu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (R.I.); (F.R.); (I.L.); (A.C.)
| | - Gavino Carboni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Roberta Loddo
- Department of Biomedical Sciences, Cittadella Universitaria Monserrato, University of Cagliari, 09042 Monserrato, Italy;
- Correspondence: (R.L.); (S.P.)
| | - Sandra Piras
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (R.I.); (F.R.); (I.L.); (A.C.)
- Correspondence: (R.L.); (S.P.)
| | - Antonio Carta
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (R.I.); (F.R.); (I.L.); (A.C.)
| |
Collapse
|
4
|
Newcomer BW. 75 years of bovine viral diarrhea virus: Current status and future applications of the use of directed antivirals. Antiviral Res 2021; 196:105205. [PMID: 34742739 DOI: 10.1016/j.antiviral.2021.105205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 01/03/2023]
Abstract
Bovine viral diarrhea virus (BVDV) was first reported 75 years ago and remains a source of major financial and production losses in the North American cattle industry. Currently, control methods in North America primarily center around biosecurity and vaccination programs; however, despite high levels of vaccination, the virus persists in the cattle herd due at least in part to the often-insidious nature of disease and the constant viremia and viral shedding of persistently infected animals which act as a reservoir for the virus. Continued development of targeted antivirals represents an additional tool for the prevention of BVDV-associated losses. Currently, in vivo studies of BVDV antivirals are relatively limited and have primarily been directed at the RNA-dependent RNA polymerase which represents the viral target with the highest potential for commercial development. Additional live animal studies have explored the potential of exogenous interferon treatment. Future research of commercial antivirals must focus on the establishment and validation of in vivo efficacy for compounds with demonstrated antiviral potential. The areas which provide the most viable economic justification for the research and development of antivirals drugs are the fed cattle sector, outbreak control, and wildlife or animals of high genetic value. With further development, targeted antivirals represent an additional tool for the management and control of BVDV in North American cattle herds.
Collapse
Affiliation(s)
- Benjamin W Newcomer
- Veterinary Education, Research, & Outreach Program, Texas A&M and West Texas A&M Universities, Canyon, TX, 79016, USA.
| |
Collapse
|
5
|
Yang G, Zhang J, Wang S, Wang J, Wang J, Zhu Y, Wang J. Gypenoside Inhibits Bovine Viral Diarrhea Virus Replication by Interfering with Viral Attachment and Internalization and Activating Apoptosis of Infected Cells. Viruses 2021; 13:v13091810. [PMID: 34578391 PMCID: PMC8473207 DOI: 10.3390/v13091810] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/18/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) causes a severe threat to the cattle industry due to ineffective control measures. Gypenoside is the primary component of Gynostemma pentaphyllum, which has potential medicinal value and has been widely applied as a food additive and herbal supplement. However, little is known about the antiviral effects of gypenoside. The present study aimed to explore the antiviral activities of gypenoside against BVDV infection. The inhibitory activity of gypenoside against BVDV was assessed by using virus titration and performing Western blotting, quantitative reverse transcription PCR (RT-qPCR), and immunofluorescence assays in MDBK cells. We found that gypenoside exhibited high anti-BVDV activity by interfering with the viral attachment to and internalization in cells. The study showed that BVDV infection inhibits apoptosis of infected cells from escaping the innate defense of host cells. Our data further demonstrated that gypenoside inhibited BVDV infection by electively activating the apoptosis of BVDV-infected cells for execution, as evidenced by the regulation of the expression of the apoptosis-related protein, promotion of caspase-3 activation, and display of positive TUNEL staining; no toxicity was observed in non-infected cells. Collectively, the data identified that gypenoside exerts an anti-BVDV-infection role by inhibiting viral attachment and internalization and selectively purging virally infected cells. Therefore, our study will contribute to the development of a novel prophylactic and therapeutic strategy against BVDV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jiufeng Wang
- Correspondence: ; Tel.: +86-010-6273-1094; Fax: +86-010-6273-1274
| |
Collapse
|
6
|
Devnarain N, Waddad AY, de la Torre BG, Albericio F, Govender T. Novel Biomimetic Human TLR2-Derived Peptides for Potential Targeting of Lipoteichoic Acid: An In Silico Assessment. Biomedicines 2021; 9:biomedicines9081063. [PMID: 34440267 PMCID: PMC8391229 DOI: 10.3390/biomedicines9081063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 01/20/2023] Open
Abstract
Antimicrobial resistance is one of the most significant threats to health and economy around the globe and has been compounded by the emergence of COVID-19, raising important consequences for antimicrobial resistance development. Contrary to conventional targeting approaches, the use of biomimetic application via nanoparticles for enhanced cellular targeting, cell penetration and localized antibiotic delivery has been highlighted as a superior approach to identify novel targeting ligands for combatting antimicrobial resistance. Gram-positive bacterial cell walls contain lipoteichoic acid (LTA), which binds specifically to Toll-like receptor 2 (TLR2) on human macrophages. This phenomenon has the potential to be exploited for the design of biomimetic peptides for antibacterial application. In this study, we have derived peptides from sequences present in human TLR2 that bind to LTA with high affinity. In silico approaches including molecular modelling, molecular docking, molecular dynamics, and thermodynamics have enabled the identification of these crucial binding amino acids, the design of four novel biomimetic TLR2-derived peptides and their LTA binding potential. The outcomes of this study have revealed that one of these novel peptides binds to LTA more strongly and stably than the other three peptides and has the potential to enhance LTA targeting and bacterial cell penetration.
Collapse
Affiliation(s)
- Nikita Devnarain
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| | - Ayman Y. Waddad
- Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa;
- Correspondence: (A.Y.W.); (T.G.); Tel.: +27-31-260-7367 (A.Y.W.); +27-31-260-7357 (T.G.)
| | - Beatriz G. de la Torre
- KRISP, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Fernando Albericio
- Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
- Correspondence: (A.Y.W.); (T.G.); Tel.: +27-31-260-7367 (A.Y.W.); +27-31-260-7357 (T.G.)
| |
Collapse
|
7
|
Yang F, Lin S, Ye F, Yang J, Qi J, Chen Z, Lin X, Wang J, Yue D, Cheng Y, Chen Z, Chen H, You Y, Zhang Z, Yang Y, Yang M, Sun H, Li Y, Cao Y, Yang S, Wei Y, Gao GF, Lu G. Structural Analysis of Rabies Virus Glycoprotein Reveals pH-Dependent Conformational Changes and Interactions with a Neutralizing Antibody. Cell Host Microbe 2020; 27:441-453.e7. [DOI: 10.1016/j.chom.2019.12.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/06/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
|
8
|
Leal ES, Adler NS, Fernández GA, Gebhard LG, Battini L, Aucar MG, Videla M, Monge ME, Hernández de Los Ríos A, Acosta Dávila JA, Morell ML, Cordo SM, García CC, Gamarnik AV, Cavasotto CN, Bollini M. De novo design approaches targeting an envelope protein pocket to identify small molecules against dengue virus. Eur J Med Chem 2019; 182:111628. [PMID: 31472473 DOI: 10.1016/j.ejmech.2019.111628] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/03/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Dengue fever is a mosquito-borne viral disease that has become a major public health concern worldwide. This disease presents with a wide range of clinical manifestations, from a mild cold-like illness to the more serious hemorrhagic dengue fever and dengue shock syndrome. Currently, neither an approved drug nor an effective vaccine for the treatment are available to fight the disease. The envelope protein (E) is a major component of the virion surface. This protein plays a key role during the viral entry process, constituting an attractive target for the development of antiviral drugs. The crystal structure of the E protein reveals the existence of a hydrophobic pocket occupied by the detergent n-octyl-β-d-glucoside (β-OG). This pocket lies at the hinge region between domains I and II and is important for the low pH-triggered conformational rearrangement required for the fusion of the virion with the host's cell. Aiming at the design of novel molecules which bind to E and act as virus entry inhibitors, we undertook a de novo design approach by "growing" molecules inside the hydrophobic site (β-OG). From more than 240000 small-molecules generated, the 2,4 pyrimidine scaffold was selected as the best candidate, from which one synthesized compound displayed micromolar activity. Molecular dynamics-based optimization was performed on this hit, and thirty derivatives were designed in silico, synthesized and evaluated on their capacity to inhibit dengue virus entry into the host cell. Four compounds were found to be potent antiviral compounds in the low-micromolar range. The assessment of drug-like physicochemical and in vitro pharmacokinetic properties revealed that compounds 3e and 3h presented acceptable solubility values and were stable in mouse plasma, simulated gastric fluid, simulated intestinal fluid, and phosphate buffered saline solution.
Collapse
Affiliation(s)
- Emilse S Leal
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia S Adler
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina; Computational Drug Design and Molecular Informatics Laboratory, Translational Medicine Research Institute (IIMT), CONICET-Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina
| | - Gabriela A Fernández
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina
| | - Leopoldo G Gebhard
- CONICET-Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, B1876, Bernal, Buenos Aires, Argentina
| | - Leandro Battini
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria G Aucar
- Computational Drug Design and Molecular Informatics Laboratory, Translational Medicine Research Institute (IIMT), CONICET-Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina
| | - Mariela Videla
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Eugenia Monge
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina
| | - Alejandro Hernández de Los Ríos
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Estrategias Antivirales, CONICET, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - John Alejandro Acosta Dávila
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Estrategias Antivirales, CONICET, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - María L Morell
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Estrategias Antivirales, CONICET, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Sandra M Cordo
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Estrategias Antivirales, CONICET, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Cybele C García
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Estrategias Antivirales, CONICET, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Andrea V Gamarnik
- Fundación Instituto Leloir-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina Buenos Aires, Argentina
| | - Claudio N Cavasotto
- Computational Drug Design and Molecular Informatics Laboratory, Translational Medicine Research Institute (IIMT), CONICET-Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina; Facultad de Ciencias Biomédicas, y Facultad de Ingeniería, Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina; Austral Institute for Artificial Intelligence, Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina
| | - Mariela Bollini
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz, 2390, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
9
|
Slater O, Kontoyianni M. The compromise of virtual screening and its impact on drug discovery. Expert Opin Drug Discov 2019; 14:619-637. [PMID: 31025886 DOI: 10.1080/17460441.2019.1604677] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Docking and structure-based virtual screening (VS) have been standard approaches in structure-based design for over two decades. However, our understanding of the limitations, potential, and strength of these techniques has enhanced, raising expectations. Areas covered: Based on a survey of reports in the past five years, we assess whether VS: (1) predicts binding poses in agreement with crystallographic data (when available); (2) is a superior screening tool, as often claimed; (3) is successful in identifying chemical scaffolds that can be starting points for subsequent lead optimization cycles. Data shows that knowledge of the target and its chemotypes in postprocessing lead to viable hits in early drug discovery endeavors. Expert opinion: VS is capable of accurate placements in the pocket for the most part, but does not consistently score screening collections accurately. What matters is capitalization on available resources to get closer to a viable lead or optimizable series. Integration of approaches, subjective hit selection guided by knowledge of the receptor or endogenous ligand, libraries driven by experimental guides, validation studies to identify the best docking/scoring that reproduces experimental findings, constraints regarding receptor-ligand interactions, thoroughly designed methodologies, and predefined cutoff scoring criteria strengthen VS's position in pharmaceutical research.
Collapse
Affiliation(s)
- Olivia Slater
- a Department of Pharmaceutical Sciences , Southern Illinois University Edwardsville , Edwardsville , IL , USA
| | - Maria Kontoyianni
- a Department of Pharmaceutical Sciences , Southern Illinois University Edwardsville , Edwardsville , IL , USA
| |
Collapse
|
10
|
Castro EF, Casal JJ, de Marco MJE, Battini L, Fabiani M, Fernández GA, Bruno AM, Cavallaro LV, Bollini M. Identification of potent bovine viral diarrhea virus inhibitors by a structure-based virtual screening approach. Bioorg Med Chem Lett 2019; 29:262-266. [PMID: 30501966 DOI: 10.1016/j.bmcl.2018.11.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/08/2018] [Accepted: 11/21/2018] [Indexed: 10/27/2022]
Abstract
Bovine viral diarrhea virus (BVDV) is a pestivirus whose infection in cattle is globally distributed. The use of antivirals could complement vaccination as a tool of control and reduce economic losses. The RNA-dependent RNA polymerase (RdRp) of the virus is essential for its genome replication and constitutes an attractive target for the identification of antivirals. With the aim of obtaining selective BVDV inhibitors, the crystal structure of BVDV RdRp was used to perform a virtual screening. Approximately 15,000 small molecules from commercial and in-house databases were evaluated and several structurally different compounds were tested in vitro for antiviral activity. Interestingly, of twelve evaluated compounds, five were active and displayed EC50 values in the sub and low-micromolar range. Time of drug addition experiment and measured intracellular BVDV RNA showed that compound 7 act during RNA synthesis. Molecular Dynamics and MM/PBSA calculation were done to characterize the interaction of the most active compounds with RdRp, which will allow future ligand optimization. These studies highlight the use of in silico screening to identify a new class of BVDV inhibitors.
Collapse
Affiliation(s)
- Eliana F Castro
- Cátedra de Virología, Departamento de Microbiología, Inmunología y Biotecnología, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan J Casal
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-CONICET, Ciudad de Buenos Aires, Argentina
| | - María J España de Marco
- Cátedra de Virología, Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Leandro Battini
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-CONICET, Ciudad de Buenos Aires, Argentina
| | - Matías Fabiani
- Cátedra de Virología, Departamento de Microbiología, Inmunología y Biotecnología, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela A Fernández
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-CONICET, Ciudad de Buenos Aires, Argentina
| | - Ana M Bruno
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Orgánica, Junín 956, C1113AAD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucía V Cavallaro
- Cátedra de Virología, Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Mariela Bollini
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-CONICET, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
11
|
Imrie F, Bradley AR, van der Schaar M, Deane CM. Protein Family-Specific Models Using Deep Neural Networks and Transfer Learning Improve Virtual Screening and Highlight the Need for More Data. J Chem Inf Model 2018; 58:2319-2330. [DOI: 10.1021/acs.jcim.8b00350] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Fergus Imrie
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, U.K
| | - Anthony R. Bradley
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, U.K
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, U.K
- Diamond Light Source Ltd., Didcot OX11 0DE, U.K
| | - Mihaela van der Schaar
- Department of Engineering, University of Oxford, Oxford OX1 3PJ, U.K
- Alan Turing Institute, London NW1 2DB, U.K
| | - Charlotte M. Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, U.K
| |
Collapse
|
12
|
Siramshetty VB, Preissner R, Gohlke BO. Exploring Activity Profiles of PAINS and Their Structural Context in Target–Ligand Complexes. J Chem Inf Model 2018; 58:1847-1857. [DOI: 10.1021/acs.jcim.8b00385] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vishal B. Siramshetty
- Structural Bioinformatics Group, Charité-Universitätsmedizin Berlin, 10115 Berlin, Germany
- BB3R - Berlin Brandenburg 3R Graduate School, Freie Universität Berlin, 14195 Berlin, Germany
| | - Robert Preissner
- Structural Bioinformatics Group, Charité-Universitätsmedizin Berlin, 10115 Berlin, Germany
- BB3R - Berlin Brandenburg 3R Graduate School, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bjoern-Oliver Gohlke
- Structural Bioinformatics Group, Charité-Universitätsmedizin Berlin, 10115 Berlin, Germany
| |
Collapse
|