1
|
Mishra S, Garg P, Trivedi M, Srivastava P. Multiple system biology approaches reveals the role of the hsa-miR-21 in increasing risk of neurological disorders in patients suffering from hypertension. J Hum Hypertens 2025:10.1038/s41371-025-01027-3. [PMID: 40389629 DOI: 10.1038/s41371-025-01027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 04/11/2025] [Accepted: 05/02/2025] [Indexed: 05/21/2025]
Abstract
Hypertension is a prevalent disease that substantially elevates the risk of neurological disorders such as dementia, stroke and Parkinson's disease. MicroRNAs (miRNAs) play a critical role in the regulation of gene expression related to brain function and disorders. Understanding the involvement of miRNAs in these conditions could provide new insights into potential therapeutic targets. The main objective of this study is to target and investigate microRNAs (miRNAs) associated with neurological disorders in patients suffering from hypertension. The genes involved in hypertension were identified from various databases including GeneCard, MalaCard, DisGeNet, OMIM & GEO2R. The key gene for hypertension was identified using a systems biology approach. Also, potent phytochemical for hypertension was determined by computer-aided drug-designing approach. Functional miRNAs were determined for the key target gene using miRNet analytics platform by hypergeometric tests. Further, the gene-miRNA interaction was determined and enrichment analysis was done. RPS27A was identified as a key target gene for hypertension. Naringenin showed effective molecular interaction with RPS27A with a binding energy score (-6.28). Further, a list of miRNAs which were targeting brain disorders was determined from miRNet. A gene-miRNA network was constructed using the PSRR tool for Parkinson's Disease, Autism Spectrum Disorder, Acute Cerebral Infarction, ACTH-Secreting Pituitary Adenoma, & Ependymoma. Further, miRNA 21 & miRNA 16 were found to be associated with four of the neurological disorders. The study identifies specific miRNAs that may serve as potential biomarkers for brain disorders in hypertensive patients. Targeting these miRNAs could open new avenues for therapeutic strategies aimed at mitigating neurological damage in this patient population.
Collapse
Affiliation(s)
- Sanjana Mishra
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Prekshi Garg
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Mala Trivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, India.
| | - Prachi Srivastava
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, India.
| |
Collapse
|
2
|
Rapún J, Pérez-Martín S, Cámara-Checa A, San José G, Núñez-Fernández R, Crespo-García T, Hoban A, Rubio-Alarcón M, Martínez-Blanco E, Tamargo J, Díez-Guerra FJ, López B, Gómez R, González A, Delpón E, Caballero R. Two concurrent mechanisms are responsible for the I Na increase produced by dapagliflozin and empagliflozin in healthy and heart failure cardiomyocytes. Biomed Pharmacother 2025; 186:117984. [PMID: 40101587 DOI: 10.1016/j.biopha.2025.117984] [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: 12/19/2024] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Dapagliflozin and empagliflozin exert many cardiovascular protective actions in heart failure (HF) patients. HF-induced electrical remodelling decreases the expression of Nav1.5 channels (encoded by SCN5A) that generate the cardiac Na+ current (INa) impairing excitability and promoting arrhythmias. We aimed to mechanistically decipher the peak INa increase produced by dapagliflozin and empagliflozin in healthy and HF cardiomyocytes. We recorded macroscopic and single-channel currents and action potentials (AP) using the patch-clamp technique and generated a mouse model of HF with reduced ejection fraction by transverse aortic constriction (TAC). Single-channel recordings showed that dapagliflozin and empagliflozin (1 μM) increased the open probability (Po) of Nav1.5 channels by augmenting channel re-openings and the number of traces with openings and by doubling the open time constant, respectively. Both drugs increased SCN5A mRNA levels and the membrane expression of Nav1.5 channels. Empagliflozin also enhanced the cytoplasmic mobility of Nav1.5 channels. Molecular modelling and site-directed mutagenesis analysis demonstrated that both drugs bind to a previously unknown site at the Nav1.5 DIII-DIV fenestration. Dapagliflozin and empagliflozin hyperpolarized the resting membrane potential and increased the action potential amplitude in human cardiomyocytes derived from induced pluripotent stem cells. Importantly, in TAC cardiomyocytes dapagliflozin and empagliflozin restored the HF-reduced peak INa to control levels. Dapagliflozin and empagliflozin bind to a novel site within cardiac Nav1.5 increasing INa by augmenting the Po and the membrane expression of the channels. We hypothesized that this unique effects could be of interest for the treatment of arrhythmias associated with decreased Nav1.5 channel expression.
Collapse
Affiliation(s)
- Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Sara Pérez-Martín
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Gorka San José
- CIBERCV, Instituto de Salud Carlos III, Spain; Program of Cardiovascular Disease, CIMA Universidad de Navarra and IdiSNA, Pamplona 31008, Spain
| | - Roberto Núñez-Fernández
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Adam Hoban
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - Elena Martínez-Blanco
- Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| | - F Javier Díez-Guerra
- Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Begoña López
- CIBERCV, Instituto de Salud Carlos III, Spain; Program of Cardiovascular Disease, CIMA Universidad de Navarra and IdiSNA, Pamplona 31008, Spain
| | - Ricardo Gómez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain
| | - Arantxa González
- CIBERCV, Instituto de Salud Carlos III, Spain; Program of Cardiovascular Disease, CIMA Universidad de Navarra and IdiSNA, Pamplona 31008, Spain; Department of Pathology, Anatomy and Physiology, Universidad de Navarra, Pamplona 31009, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain.
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid 28040, Spain; CIBERCV, Instituto de Salud Carlos III, Spain
| |
Collapse
|
3
|
Çankaya N, Kebiroğlu MH, Yalcin Azarkan S. Computational investigation of m-acetamide and 3MPAEA: Characterization, toxicity, and molecular docking and dynamic analyses. Drug Chem Toxicol 2025:1-16. [PMID: 40293256 DOI: 10.1080/01480545.2025.2496358] [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: 01/28/2025] [Revised: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
In this study, 2-(3-methoxyphenylamino)-2-oxoethyl acrylate (3MPAEA) molecule was synthesized in two steps. In the first step, 2-chloro-N-(3-methoxyphenyl)acetamide (m-acetamide) was obtained. Density functional theory (DFT) calculations were performed to obtain information about the electronic and structural properties of the synthesized molecules. The Raman Spectrum and UV-Visible analysis were calculated using the Gaussian package program. Additionally, Natural Bond Orbital (NBO) Analysis, Electron Localization Function (ELF), Electrostatic Potential Map (ESP), Average Local Ionization Energy (ALIE), and the toxicological properties of the molecules were examined. Simultaneously, molecular docking and dynamic analyses were conducted to investigate the interaction of m-acetamide and 3MPAEA with proteins involved in nuclear receptor signaling pathways, stress response pathways, molecular initiating events, and metabolism, as identified in the protox analysis. The findings aligned with the protox analysis results. The results obtained provide new insights into the electronic and toxicological properties of these molecules.
Collapse
Affiliation(s)
- Nevin Çankaya
- Vocational School of Health Services, Usak University, Usak, Turkey
| | - Mehmet Hanifi Kebiroğlu
- Darende Bekir Ilicak Vocational School, Department of Opticianry, Malatya Turgut Ozal University, Malatya, Turkey
| | - Serap Yalcin Azarkan
- Department of Medical Pharmacology, Faculty of Medicine, Kırsehir Ahi Evran University, Kırşehir, Turkey
| |
Collapse
|
4
|
Ianeselli A, Howard J, Gerstein MB. A discard-and-restart MD algorithm for the sampling of protein intermediate states. Biophys J 2025:S0006-3495(25)00198-5. [PMID: 40156184 DOI: 10.1016/j.bpj.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/20/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025] Open
Abstract
We introduce a discard-and-restart molecular dynamics (MD) algorithm tailored for the sampling of realistic protein intermediate states. It aids computational structure-based drug discovery by reducing the simulation times to compute a "quick sketch" of folding pathways by up to 2000×. The algorithm iteratively performs short MD simulations and measures their proximity to a target state via a collective variable loss, which can be defined in a flexible fashion, locally or globally. Using the loss, if the trajectory proceeds toward the target, the MD simulation continues. Otherwise, it is discarded, and a new MD simulation is restarted, with new initial velocities randomly drawn from a Maxwell-Boltzmann distribution. The discard-and-restart algorithm demonstrates efficacy and atomistic accuracy in capturing the folding pathways in several contexts: 1) fast-folding small protein domains, 2) the folding intermediate of the prion protein PrP, and 3) the spontaneous partial unfolding of α-tubulin, a crucial event for microtubule severing. During each iteration of the algorithm, we can perform AI-based analysis of the transitory conformations to find potential binding pockets, which could represent druggable sites. Overall, our algorithm enables systematic and computationally efficient exploration of conformational landscapes, enhancing the design of ligands targeting dynamic protein states.
Collapse
Affiliation(s)
- Alan Ianeselli
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Mark B Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut; Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut; Department of Computer Science, Yale University, New Haven, Connecticut; Department of Statistics and Data Science, Yale University, New Haven, Connecticut; Department of Biomedical Informatics & Data Science, Yale University, New Haven, Connecticut.
| |
Collapse
|
5
|
da Conceição PJP, Ayusso GM, Carvalho T, Duarte Lima ML, Marinho MDS, Moraes FR, Galán-Jurado PE, González-Santamaría J, Bittar C, Zhang B, Jardim ACG, Rahal P, Calmon MF. In Vitro Evaluation of the Antiviral Activity of Polyphenol (-)-Epigallocatechin-3-Gallate (EGCG) Against Mayaro Virus. Viruses 2025; 17:258. [PMID: 40007013 PMCID: PMC11860591 DOI: 10.3390/v17020258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The Mayaro virus (MAYV), Togaviridae family, genus Alphavirus, has caused several sporadic outbreaks, affecting countries in the Americas. Currently, there are no licensed drugs against MAYV, requiring the search for effective antiviral compounds. Thus, this study aimed to evaluate the antiviral potential of polyphenol (-)-epigallocatechin-3-gallate (EGCG) against MAYV infection, in vitro. Antiviral assays against MAYV were performed in BHK-21 and Vero E6 cells. In addition, molecular docking was performed with EGCG and the MAYV non-structural and structural proteins. EGCG showed a significant protective effect against MAYV infection in both cell lines. The virucidal assay showed an effect on extracellular viral particles at the entry stage into BHK-21 cells. Finally, it also showed significant inhibition in the post-entry stages of the MAYV replication cycle, acting on the replication of the genetic material and late stages, such as assembly and release. In addition, the MAYV proteins E1 and nsP1 were significantly inhibited by the EGCG treatment in BHK-21 cells. Molecular docking analysis also showed that EGCG could interact with MAYV Capsid and Envelope proteins (E1 and E2). Therefore, this study shows the potential of EGCG as a promising antiviral against MAYV, as it acts on different stages of the MAYV replication cycle.
Collapse
Affiliation(s)
- Pâmela Jóyce Previdelli da Conceição
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (P.J.P.d.C.); (G.M.A.); (M.L.D.L.); (P.R.)
| | - Gabriela Miranda Ayusso
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (P.J.P.d.C.); (G.M.A.); (M.L.D.L.); (P.R.)
| | - Tamara Carvalho
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique (CNRS), 34000 Montpellier, France;
| | - Maria Leticia Duarte Lima
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (P.J.P.d.C.); (G.M.A.); (M.L.D.L.); (P.R.)
| | - Mikaela dos Santos Marinho
- Laboratory of Antiviral Research, Institute of Biomedical Science, ICBIM/UFU, Uberlândia 38405-302, MG, Brazil; (M.d.S.M.); (A.C.G.J.)
| | - Fábio Rogério Moraes
- Physics Department, São Paulo State University—UNESP, São José do Rio Preto 15385-000, SP, Brazil;
| | - Paola Elaine Galán-Jurado
- Grupo de Biología Celular y Molecular de Arbovirus, Departamento de Genómica y Proteómica, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panamá City 0816-02593, Panama; (P.E.G.-J.); (J.G.-S.)
| | - José González-Santamaría
- Grupo de Biología Celular y Molecular de Arbovirus, Departamento de Genómica y Proteómica, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panamá City 0816-02593, Panama; (P.E.G.-J.); (J.G.-S.)
| | - Cíntia Bittar
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA;
| | - Bo Zhang
- Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
| | - Ana Carolina Gomes Jardim
- Laboratory of Antiviral Research, Institute of Biomedical Science, ICBIM/UFU, Uberlândia 38405-302, MG, Brazil; (M.d.S.M.); (A.C.G.J.)
| | - Paula Rahal
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (P.J.P.d.C.); (G.M.A.); (M.L.D.L.); (P.R.)
| | - Marilia Freitas Calmon
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (P.J.P.d.C.); (G.M.A.); (M.L.D.L.); (P.R.)
| |
Collapse
|
6
|
Takayama I, Araki N, Tewari J, Yohda M, Matsunami H, Fukutani Y. Antagonists Enhance Cell-Surface Expression of Mammalian Odorant Receptors. Int J Mol Sci 2025; 26:1458. [PMID: 40003926 PMCID: PMC11855683 DOI: 10.3390/ijms26041458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Functional characterization of vertebrate odorant receptors (ORs), members of the G protein-coupled receptor (GPCR) family, is essential for understanding olfaction. However, the functional expression of ORs in heterologous cells is often challenging, at least partly caused by structural instability in non-olfactory cells. Antagonists have been shown to restore membrane expression of some non-olfactory GPCR mutants, likely by transient increase in structural stability upon antagonist binding. Based on this premise, we examined whether antagonists could enhance OR membrane expression in heterologous cells. Using phenyl salicylate (PES) on cells expressing the mouse OR Or11g7, we observed increased cell surface expression exceeding the effects of co-expression with the OR chaperone RTP1S. After removing the antagonist, Or11g7 retained normal agonist responsiveness. Similar enhancements in cell surface expression were observed for a human OR OR2T11 treated with its antagonists. These findings suggest that small-molecule antagonists act as pharmacological chaperones to stabilize OR conformation, enhancing surface expression in a manner similar to molecular chaperones. Our study reveals a novel role for odorant antagonists in OR biogenesis and may inform future research on olfactory training mechanisms.
Collapse
Affiliation(s)
- Ikumi Takayama
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei 184-8588, Tokyo, Japan; (I.T.); (N.A.); (M.Y.)
| | - Nako Araki
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei 184-8588, Tokyo, Japan; (I.T.); (N.A.); (M.Y.)
| | - Jeevan Tewari
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Masafumi Yohda
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei 184-8588, Tokyo, Japan; (I.T.); (N.A.); (M.Y.)
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yosuke Fukutani
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei 184-8588, Tokyo, Japan; (I.T.); (N.A.); (M.Y.)
| |
Collapse
|
7
|
Petrovic D, Slade L, Paikopoulos Y, D'Andrea D, Savic N, Stancic A, Miljkovic JL, Vignane T, Drekolia MK, Mladenovic D, Sutulovic N, Refeyton A, Kolakovic M, Jovanovic VM, Zivanovic J, Miler M, Vellecco V, Brancaleone V, Bucci M, Casey AM, Yu C, Kasarla SS, Smith KW, Kalfe-Yildiz A, Stenzel M, Miranda-Vizuete A, Hergenröder R, Phapale P, Stanojlovic O, Ivanovic-Burmazovic I, Vlaski-Lafarge M, Bibli SI, Murphy MP, Otasevic V, Filipovic MR. Ergothioneine improves healthspan of aged animals by enhancing cGPDH activity through CSE-dependent persulfidation. Cell Metab 2025; 37:542-556.e14. [PMID: 39842434 DOI: 10.1016/j.cmet.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/22/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025]
Abstract
Ergothioneine (ET), a dietary thione/thiol, is receiving growing attention for its possible benefits in healthy aging and metabolic resilience. Our study investigates ET's effects on healthspan in aged animals, revealing lifespan extension and enhanced mobility in Caenorhabditis elegans, accompanied by improved stress resistance and reduced age-associated biomarkers. In aged rats, ET administration enhances exercise endurance, muscle mass, and vascularization, concomitant with higher NAD+ levels in muscle. Mechanistically, ET acts as an alternative substrate for cystathionine gamma-lyase (CSE), stimulating H2S production, which increases protein persulfidation of more than 300 protein targets. Among these, protein-persulfidation-driven activation of cytosolic glycerol-3-phosphate dehydrogenase (cGPDH) primarily contributes to the ET-induced NAD+ increase. ET's effects are abolished in models lacking CSE or cGPDH, highlighting the essential role of H2S signaling and protein persulfidation. These findings elucidate ET's multifaceted actions and provide insights into its therapeutic potential for combating age-related muscle decline and metabolic perturbations.
Collapse
Affiliation(s)
- Dunja Petrovic
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Luke Slade
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | | | - Davide D'Andrea
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Nevena Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Stancic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jan Lj Miljkovic
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK
| | - Thibaut Vignane
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Maria Kyriaki Drekolia
- Department of Vascular Dysfunction, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dusan Mladenovic
- Institute for Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Sutulovic
- Laboratory for Neurophysiology, Institute for Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Alice Refeyton
- Inserm U1211 Maladies Rares: Génétique et Métabolisme, Université de Bordeaux, Bordeaux, France
| | - Milica Kolakovic
- Department of Chemistry, Ludwig Maximilians University of Munich, Munich, Germany
| | - Vladimir M Jovanovic
- Bioinformatics Solution Center, Institute for Informatics, Freie Universität Berlin, Berlin, Germany
| | - Jasmina Zivanovic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marko Miler
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Alva M Casey
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK
| | - ChakShun Yu
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK
| | | | | | | | - Martin Stenzel
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Antonio Miranda-Vizuete
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | | | - Prasad Phapale
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Olivera Stanojlovic
- Laboratory for Neurophysiology, Institute for Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Marija Vlaski-Lafarge
- Inserm U1211 Maladies Rares: Génétique et Métabolisme, Université de Bordeaux, Bordeaux, France
| | - Sofia-Iris Bibli
- Department of Vascular Dysfunction, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Vesna Otasevic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milos R Filipovic
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany; School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
8
|
Brahimi F, Nassour H, Galan A, Guruswamy R, Ortiz C, Nejatie A, Nedev H, Trempe JF, Saragovi HU. Selective inhibitors of the TrkC.T1 receptor reduce retinal inflammation and delay neuronal death in a model of retinitis pigmentosa. PNAS NEXUS 2025; 4:pgaf020. [PMID: 39911316 PMCID: PMC11795507 DOI: 10.1093/pnasnexus/pgaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025]
Abstract
The heterogeneity of receptor isoforms can cause an apparent paradox where each isoform can promote different or even opposite biological pathways. One example is the neurotrophin receptor TrkC. The trkC mRNA translates a full-length receptor tyrosine kinase (TrkC-FL) whose activation by the growth factor NT3 promotes neuronal survival. In some diseases, the trkC mRNA is spliced to a kinase-truncated isoform (TrkC.T1) whose activation by NT3 up-regulates tumor necrosis factor alpha (TNF-α) causing neurotoxicity. Since TrkC.T1 expression is significantly increased at the onset of neurodegeneration, we hypothesized that in disease TrkC.T1-mediated toxicity prevails over TrkC-FL-mediated survival. To study this, we developed small molecules that selectively antagonize NT3-driven TrkC.T1 neurotoxicity without compromising TrkC-FL survival. In a genetic mouse model of retinitis pigmentosa, therapeutic administration of TrkC.T1 antagonists prevents elevation of TNF-α and reduces photoreceptor neuronal death. This work demonstrates the importance of accounting for functional and structural heterogeneity in receptor-ligand interactions, illustrates chemical biology strategies to develop isoform-selective agents, validates TrkC.T1 as a druggable target, and expands the therapeutic concept of reducing neurotoxicity as a strategy to achieve neuroprotection.
Collapse
Affiliation(s)
- Fouad Brahimi
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
| | - Hassan Nassour
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada H3G 1Y6
| | - Alba Galan
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
| | - Revathy Guruswamy
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada H3G 1Y6
| | - Christina Ortiz
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
| | - Ali Nejatie
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada H3G 1Y6
| | - Hinyu Nedev
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
| | - Jean-Francois Trempe
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada H3G 1Y6
- Centre de Recherche en Biologie Structurale, McGill University, Montreal, QC, Canada H3G 1Y6
- Structural Genomics Consortium, McGill University, Montreal, QC, Canada H3G 1Y6
- Brain Repair and Integrative Neuroscience (BRaIN), McGill University, Montreal, QC, Canada H3G 1Y6
| | - H Uri Saragovi
- Lady Davis Institute—Jewish General Hospital, McGill University, Center for Translational Research, Montreal, QC, Canada H3T 1E2
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada H3G 1Y6
- Brain Repair and Integrative Neuroscience (BRaIN), McGill University, Montreal, QC, Canada H3G 1Y6
- Ophthalmology and Vision Science, McGill University, Montreal, QC, Canada H3T 1E2
| |
Collapse
|
9
|
Basmenj ER, Pajhouh SR, Ebrahimi Fallah A, naijian R, Rahimi E, Atighy H, Ghiabi S, Ghiabi S. Computational epitope-based vaccine design with bioinformatics approach; a review. Heliyon 2025; 11:e41714. [PMID: 39866399 PMCID: PMC11761309 DOI: 10.1016/j.heliyon.2025.e41714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
The significance of vaccine development has gained heightened importance in light of the COVID-19 pandemic. In such critical circumstances, global citizens anticipate researchers in this field to swiftly identify a vaccine candidate to combat the pandemic's root cause. It is widely recognized that the vaccine design process is traditionally both time-consuming and costly. However, a specialized subfield within bioinformatics, known as "multi-epitope vaccine design" or "reverse vaccinology," has significantly decreased the time and costs of the vaccine design process. The methodology reverses itself in this subfield and finds a potential vaccine candidate by analyzing the pathogen's genome. Leveraging the tools available in this domain, we strive to pinpoint the most suitable antigen for crafting a vaccine against our target. Once the optimal antigen is identified, the next step involves uncovering epitopes within this antigen. The immune system recognizes particular areas of an antigen as epitopes. By characterizing these crucial segments, we gain the opportunity to design a vaccine centered around these epitopes. Subsequently, after identifying and assembling the vital epitopes with the assistance of linkers and adjuvants, our vaccine candidate can be formulated. Finally, employing computational techniques, we can thoroughly evaluate the designed vaccine. This review article comprehensively covers the entire multi-epitope vaccine development process, starting from obtaining the pathogen's genome to identifying the relevant vaccine candidate and concluding with an evaluation. Furthermore, we will delve into the essential tools needed at each stage, comparing and introducing them.
Collapse
Affiliation(s)
| | | | | | - Rafe naijian
- Student research committee, faculty of pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Elmira Rahimi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Atighy
- School of Pharmacy, Centro Escolar University, Manila, Philippines
| | - Shadan Ghiabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shamim Ghiabi
- Tehran Azad University of Medical Sciences, Faculty of Pharmaceutical Sciences, Iran
| |
Collapse
|
10
|
Nagarajan U, Naha A, Ashok G, Balasubramanian A, Ramaiah S, V Kanth S, Dusthackeer A, Anbarasu A, Natarajan S. Biomolecular Interaction of Carnosine and Anti-TB Drug: Preparation of Functional Biopeptide-Based Nanocomposites and Characterization through In Vitro and In Silico Investigations. ACS OMEGA 2025; 10:567-587. [PMID: 39829512 PMCID: PMC11739970 DOI: 10.1021/acsomega.4c07176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 01/22/2025]
Abstract
Host-directed therapies (HDTs) resolve excessive inflammation during tuberculosis (TB) disease, which leads to irreversible lung tissue damage. The peptide-based nanostructures possess intrinsic anti-inflammatory and antioxidant properties among HDTs. Native carnosine, a natural dipeptide with superior self-organization and functionalities, was chosen for nanoformulation. In the present work, multiscale self-assembly approaches of carnosine were developed using a solvent-mediated process (hexafluoro-2-propanol) and further linked with first-line anti-TB drugs. The organofluorine compound in a solvent is attributed to the self-assembling process with heteroatom acceptors in carnosine. In the carnosine-anti-TB drug nanocomposite, the functional moieties represent the involvement of hydrogen bonding and the electrostatic force of attraction. The minimum inhibitory concentration of carnosine-anti-TB drug composites represents an antimycobacterial effect on par with standard drugs. The silicon findings complemented the in vitro results through quantum chemical simulations, elucidating the respective binding pockets between putative Mtb drug targets and carnosine-anti-TB composites. These findings confirmed that the carnosine and anti-TB drug nanocomposites prepared through a solvent-mediated process act as a rational design for functional nanodelivery systems for sustainable TB therapeutics.
Collapse
Affiliation(s)
- Usharani Nagarajan
- Department
of Biochemistry, ICMR-National Institute
for Research in Tuberculosis (NIRT), Chennai 600 031, India
| | - Aniket Naha
- Department
of Bio-Medical Sciences, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632 014, India
- Medical
Biotechnology and Computational Drug Designing Laboratory, Pushpagiri
Research Centre, Pushpagiri Medical Society, Tiruvalla,Kerala 689 101, India
| | - Gayathri Ashok
- Medical
and Biological Computing Laboratory, SBST, VIT, Vellore, Tamil Nadu 632 014, India
- Department
of Biosciences, SBST, VIT, Vellore, Tamil Nadu 632 014, India
| | | | - Sudha Ramaiah
- Medical
and Biological Computing Laboratory, SBST, VIT, Vellore, Tamil Nadu 632 014, India
- Department
of Biosciences, SBST, VIT, Vellore, Tamil Nadu 632 014, India
| | - Swarna V Kanth
- CHORD, CSIR-Central Leather Research Institute, Chennai 600 020, India
| | - Azger Dusthackeer
- Department
of Bacteriology, ICMR-National Institute
for Research in Tuberculosis (NIRT), Chennai 600 031, India
| | - Anand Anbarasu
- Medical
and Biological Computing Laboratory, SBST, VIT, Vellore, Tamil Nadu 632 014, India
- Department
of Biotechnology, SBST, VIT, Vellore, Tamil Nadu 632 014, India
| | - Saravanan Natarajan
- Department
of Biochemistry, ICMR-National Institute
for Research in Tuberculosis (NIRT), Chennai 600 031, India
| |
Collapse
|
11
|
Tamturk E, Yalcın S, Ercan F, Tuncbilek AS. In vivo, In vitro, and In silico Studies of Umbelliferone and Irinotecan on MDA-MB-231 Breast Cancer Cell Line and Drosophila melanogaster Larvae. Anticancer Agents Med Chem 2025; 25:499-516. [PMID: 39473207 DOI: 10.2174/0118715206340868241018075528] [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: 07/02/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 05/07/2025]
Abstract
AIMS Deaths from cancer are still very common all over the world and continue to be the focus of scientific research. Chemotherapy is one of the primary treatments used to prevent deaths from cancer. Side effects of chemotherapeutic drugs and resistance of cells to drugs are essential problems that limit the treatment process. Drug combination therapy is regarded as a significant application that inhibits the growth of tumors and is anticipated to provide a solution for the issues encountered. The combination therapy aims at a synergistic effect that will limit drug resistance and cytotoxic effects with appropriate drug combinations. In this context, we aim to investigate the In vitro, In vivo, and In silico effects of single and combined doses of umbelliferone and irinotecan, known for their anticarcinogenic and curative effects, on MDA-MB-231 breast cancer cell lines and the model organism Drosophila melanogaster. Background: Irinotecan is currently used as an anticarcinogenic drug. Anticarcinogenic effects of umbelliferone have also been detected. The in vivo, in vitro, and in silico impacts of single and combined doses use of these two agents are not yet available in the literature. OBJECTIVE This study aims to determine the anticarcinogenic effects of single and combined use of umbelliferone and irinotecan at the molecular level. It also attempts to determine the binding energies of chemicals to cancerrelated proteins through docking and molecular dynamic studies. METHODS The cytotoxic effects of individual and combinational doses of umbelliferone and irinotecan on the MDAMB- 231 cell line and D. melanogaster were calculated by XTT and probit analyses. IC50 values for the cancer cells, LC50, and LC99 values for D. melanogaster were found. Gene expression analysis was performed to determine the effects of chemical agents on miR-7, miR-11, and miR-14, and their expression levels were found. The sequences of miRNAs not found in the literature were determined, and their molecular imaging was performed. In addition, the binding energies of irinotecan and umbelliferone to Bcl-2, Bad, and Akt1 proteins, which are known to have apoptotic effects, were found by the molecular docking method. Molecular dynamics studies of Bad proteins and chemicals were also performed. The drug potential of chemicals was determined by ADME/T analysis. RESULTS The cytotoxic effect on cells was calculated, and the IC50 value of umbelliferone was calculated as 158 μM, the IC50 value of irinotecan was calculated as 48,3 μM and the IC50 value was calculated as 20 μM. In the probit analysis performed to calculate the cytotoxic effects of drugs on D. melanogaster, the LC50 value of umbelliferone was 2,5 μM, and the LC99 value was 13,4 μM. The LC50 value of irinotecan was found to be 0,1 μM, and the LC99 value was 0,28 μM. It was concluded that single and combined doses of chemicals in the invasion experiment significantly affected the spread of cells. As a result of expression analysis, a significant increase in HsamiR- 7 (Homo sapiens miRNA-7), Hsa-miR-14 (Homo sapiens miRNA-14), and Hsa-miR-11(Homo sapiens miRNA-11) expression was observed in cells treated with umbelliferone irinotecan compared to the control groups. CONCLUSION In our study, it can be concluded that the cytotoxic effects of individual and combination doses of umbelliferone and irinotecan on MDA-MB-231 cells and D. melanogaster larvae are significant. In addition, the effects of umbelliferone and irinotecan on the expression level of miR-7, which is a common D. melanogaster and human miRNA, should be widely investigated. Expression analyses and docking studies of Hsa-miR-11 and Hsa-miR-14, which have been newly studied and are not in data repositories, are important for cancer research. In particular, the expression and binding energy of these miRNAs in new drug combinations and the expression level in different cancer cell lines are important for future studies. Another crucial point is that in vivo tests using different model species validate the usage of drugs at both single and mixed dosages. Other: As a result of this study, the In vivo, In vitro, and In silico effects of single and combined doses of umbelliferone and irinotecan were determined. In future studies, it would be useful to determine the binding energies of umbelliferone and irinotecan to other cancer-related proteins and to find their interactions with different miRNAs. Additionally, studies on different model organisms are also important.
Collapse
Affiliation(s)
- Erkut Tamturk
- Department of Biology, Faculty of Art and Sciences, Erciyes University, 38100, Kayseri, Türkiye
| | - Serap Yalcın
- Department of Medical Pharmacology, Faculty of Medicine, Kırşehir Ahi Evran University, 40100, Kırşehir, Türkiye
| | - Fahriye Ercan
- Department of Plant Protection, Faculty of Agriculture, Kırşehir Ahi Evran University, 40100, Kırşehir, Türkiye
| | - Aydın Suzu Tuncbilek
- Department of Biology, Faculty of Art and Sciences, Erciyes University, 38100, Kayseri, Türkiye
| |
Collapse
|
12
|
Uba AI. Computer-Aided Design of VEGFR-2 Inhibitors as Anticancer Agents: A Review. J Mol Recognit 2025; 38:e3104. [PMID: 39389566 DOI: 10.1002/jmr.3104] [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: 06/21/2024] [Revised: 08/01/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024]
Abstract
Due to its intricate molecular and structural characteristics, vascular endothelial growth factor receptor 2 (VEGFR-2) is essential for the development of new blood vessels in various pathological processes and conditions, especially in cancers. VEGFR-2 inhibitors have demonstrated significant anticancer effects by blocking many signaling pathways linked to tumor growth, metastasis, and angiogenesis. Several small compounds, including the well-tolerated sunitinib and sorafenib, have been approved as VEGFR-2 inhibitors. However, the widespread side effects linked to these VEGFR-2 inhibitors-hypertension, epistaxis, proteinuria, and upper respiratory infection-motivate researchers to search for new VEGFR-2 inhibitors with better pharmacokinetic profiles. The key molecular interactions required for the interaction of the small molecules with the protein target to produce the desired pharmacological effects are identified using computer-aided drug design (CADD) methods such as pharmacophore and QSAR modeling, structure-based virtual screening, molecular docking, molecular dynamics (MD) simulation coupled with MM/PB(GB)SA, and other computational strategies. This review discusses the applications of these methods for VEGFR-2 inhibitor design. Future VEGFR-2 inhibitor designs may be influenced by this review, which focuses on the current trends of using multiple screening layers to design better inhibitors.
Collapse
Affiliation(s)
- Abdullahi Ibrahim Uba
- Department of Molecular Biology and Genetics, Istanbul AREL University, Istanbul, Turkey
| |
Collapse
|
13
|
Vempati RK, Malla RR. Coralyne Targets the Catalytic Domain of MMP9: An In Silico and In Vitro Investigation. Crit Rev Oncog 2025; 30:71-89. [PMID: 39819436 DOI: 10.1615/critrevoncog.2024056393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Coralyne (COR) is a protoberberine-like isoquinoline alkaloid, and it is known for double-stranded (ds) DNA intercalation and topoisomerase inhibition. It can also sensitize cancer cells through various mechanisms. COR reduces the proliferation and migration of breast cancer cells by inhibiting the expression and activity of matrix metalloproteinase 9 (MMP9). However, the mechanism involved in the inhibitory activity of COR on MMP9 is not known. In the present study, in silico docking studies showed that COR binds to the active site of MMP9 catalytic domain (MMP9-CD) with considerable affinity. The binding affinity of COR to the MMP9-CD, estimated by three different web servers: CB Dock, Seam Dock, and PyRx, was found to be either -7.4 or -7.5 kcal/mol. Another web server that is routinely used for docking studies, Docking Server, has predicted a binding affinity of -5.9 kcal/mol. All four docking servers predicted the same binding site for COR within the MMP9-CD. Corroborating our docking results, molecular dynamic simulation studies have also shown that COR interacts with the same key active site amino acid residues of the MMP9-CD that are essential for its proteolytic function. Molecular mechanics with generalized born and surface area (MMGBSA) calculations using Schrodinger's prime module have shown that the binding free energy with which COR binds to MMP9 is -50 kcal/mol. It inhibited activity of recombinant human MMP9 activity and induced significant cytotoxicity and reduced the proliferation of MDA-MB 468 cells. Overall, our in silico and in vitro experiments show that COR potentially inhibits the activity of MMP9 by directly binding to the active site of its catalytic domain and possibly inhibits proliferation of MDA-MB 468 cells.
Collapse
Affiliation(s)
- Rahul Kumar Vempati
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam, India
| | | |
Collapse
|
14
|
Angelia MRN, Rodelas-Angelia AJD, Yang C, Park S, Jeong SP, Jang H, Bela-ong DB, Jang H, Thompson KD, Jung T. Screening and Characterization of Sialic Acid-Binding Variable Lymphocyte Receptors from Hagfish. BIOTECH 2024; 13:46. [PMID: 39584903 PMCID: PMC11586995 DOI: 10.3390/biotech13040046] [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: 09/02/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024] Open
Abstract
Sialic acid is a diverse group of monosaccharides often found on the termini of N- and O-linked glycans as well as being components of glycoconjugates. Hypersialylation has been associated with the progression of chronic inflammation-mediated diseases such as cardiovascular disease and cancer. Given its role in infection and disease-related processes, sialic acid is a promising target for therapeutic approaches that utilize carbohydrate-binding molecules. In this study, we screened for sialic acid-recognizing variable lymphocyte receptors (VLRBs) or ccombodies from inshore hagfish (Eptatretus burgeri) using a synthetic Neu5Ac-glycoconjugate as an antigen in immunoassay. Resulting ccombodies, 2D8, 5G11, 4A1, and 5F8 were further characterized in terms of their binding activity and specificity. A competitive ELISA using free haptens showed strong inhibition using either N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc). The half-maximal inhibitory concentrations (IC50) for Neu5Ac ranged from 7.02 to 17.06 mM, with candidates 4A1 and 5G11 requiring the least and highest amounts, respectively. IC50 values for Neu5Gc ranged from 8.12 to 13.91 mM, for 4A1 and 5G11, respectively. Candidate ccombodies also detected naturally occurring sialic acid from known sialoglycoproteins using a dot blot assay. Neu5Gc-5G11 and Neu5Ac-2D8 yielded the strongest and weakest docking interactions with affinity values of -5.9 kcal/mol and -4.9 kcal/mol, respectively. Hydrogen bonding and hydrophobic interactions were predicted to be the predominant noncovalent forces observed between the ccombodies and sialic acid. This study demonstrates that glycan-binding VLRBs from hagfish hold promise in augmenting the glycobiologists' toolkit in investigating the roles of glycans in human and animal health and disease.
Collapse
Affiliation(s)
- Mark Rickard N. Angelia
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, College, Laguna 4031, Philippines
| | - Abigail Joy D. Rodelas-Angelia
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| | - Cheolung Yang
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| | - Sojeong Park
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| | - Seung pyo Jeong
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| | - Hyeok Jang
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| | - Dennis Berbulla Bela-ong
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| | - Hobin Jang
- Center for Study of Emerging and Re-Emerging Viruses, Korea Virus Research Institute, Institute for Basic Science, Daejeon 34126, Republic of Korea;
| | - Kim D. Thompson
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Midlothian EH26 0PZ, UK;
| | - Taesung Jung
- Laboratory of Aquatic Animal Diseases, Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-Daero, Jinju-si 52828, Gyeongsangnam-do, Republic of Korea; (M.R.N.A.); (A.J.D.R.-A.); (C.Y.); (S.P.); (S.p.J.); (H.J.); (D.B.B.-o.)
| |
Collapse
|
15
|
Ceres K, Zehr JD, Murrell C, Millet JK, Sun Q, McQueary HC, Horton A, Cazer C, Sams K, Reboul G, Andreopoulos WB, Mitchell PK, Anderson R, Franklin-Guild R, Cronk BD, Stanhope BJ, Burbick CR, Wolking R, Peak L, Zhang Y, McDowall R, Krishnamurthy A, Slavic D, Sekhon PK, Tyson GH, Ceric O, Stanhope MJ, Goodman LB. Evolutionary genomic analyses of canine E. coli infections identify a relic capsular locus associated with resistance to multiple classes of antimicrobials. Appl Environ Microbiol 2024; 90:e0035424. [PMID: 39012166 PMCID: PMC11337803 DOI: 10.1128/aem.00354-24] [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: 02/28/2024] [Accepted: 06/08/2024] [Indexed: 07/17/2024] Open
Abstract
Infections caused by antimicrobial-resistant Escherichia coli are the leading cause of death attributed to antimicrobial resistance (AMR) worldwide, and the known AMR mechanisms involve a range of functional proteins. Here, we employed a pan-genome wide association study (GWAS) approach on over 1,000 E. coli isolates from sick dogs collected across the US and Canada and identified a strong statistical association (empirical P < 0.01) of AMR, involving a range of antibiotics to a group 1 capsular (CPS) gene cluster. This cluster included genes under relaxed selection pressure, had several loci missing, and had pseudogenes for other key loci. Furthermore, this cluster is widespread in E. coli and Klebsiella clinical isolates across multiple host species. Earlier studies demonstrated that the octameric CPS polysaccharide export protein Wza can transmit macrolide antibiotics into the E. coli periplasm. We suggest that the CPS in question, and its highly divergent Wza, functions as an antibiotic trap, preventing antimicrobial penetration. We also highlight the high diversity of lineages circulating in dogs across all regions studied, the overlap with human lineages, and regional prevalence of resistance to multiple antimicrobial classes. IMPORTANCE Much of the human genomic epidemiology data available for E. coli mechanism discovery studies has been heavily biased toward shiga-toxin producing strains from humans and livestock. E. coli occupies many niches and produces a wide variety of other significant pathotypes, including some implicated in chronic disease. We hypothesized that since dogs tend to share similar strains with their owners and are treated with similar antibiotics, their pathogenic isolates will harbor unexplored AMR mechanisms of importance to humans as well as animals. By comparing over 1,000 genomes with in vitro antimicrobial susceptibility data from sick dogs across the US and Canada, we identified a strong multidrug resistance association with an operon that appears to have once conferred a type 1 capsule production system.
Collapse
Affiliation(s)
| | | | | | - Jean K. Millet
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, Paris, France
| | - Qi Sun
- Cornell University, Ithaca, New York, USA
| | | | | | | | - Kelly Sams
- Cornell University, Ithaca, New York, USA
| | | | | | | | | | | | | | | | - Claire R. Burbick
- Washington Animal Disease Diagnostic Laboratory, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Rebecca Wolking
- Washington Animal Disease Diagnostic Laboratory, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Laura Peak
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Yan Zhang
- Ohio Department of Agriculture Animal Disease Diagnostic Laboratory, Reynoldsburg, Ohio, USA
| | - Rebeccah McDowall
- University of Guelph, Animal Health Laboratory, Guelph, Ontario, Canada
| | | | - Durda Slavic
- University of Guelph, Animal Health Laboratory, Guelph, Ontario, Canada
| | | | - Gregory H. Tyson
- US Food and Drug Administration, Veterinary Laboratory Investigation and Response Network, Laurel, Maryland, USA
| | - Olgica Ceric
- US Food and Drug Administration, Veterinary Laboratory Investigation and Response Network, Laurel, Maryland, USA
| | | | | |
Collapse
|
16
|
Boas Lichty KE, Loughran RM, Ushijima B, Richards GP, Boyd EF. Osmotic stress response of the coral and oyster pathogen Vibrio coralliilyticus: acquisition of catabolism gene clusters for the compatible solute and signaling molecule myo-inositol. Appl Environ Microbiol 2024; 90:e0092024. [PMID: 38874337 PMCID: PMC11267925 DOI: 10.1128/aem.00920-24] [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: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024] Open
Abstract
Marine bacteria experience fluctuations in osmolarity that they must adapt to, and most bacteria respond to high osmolarity by accumulating compatible solutes also known as osmolytes. The osmotic stress response and compatible solutes used by the coral and oyster pathogen Vibrio coralliilyticus were unknown. In this study, we showed that to alleviate osmotic stress V. coralliilyticus biosynthesized glycine betaine (GB) and transported into the cell choline, GB, ectoine, dimethylglycine, and dimethylsulfoniopropionate, but not myo-inositol. Myo-inositol is a stress protectant and a signaling molecule that is biosynthesized and used by algae. Bioinformatics identified myo-inositol (iol) catabolism clusters in V. coralliilyticus and other Vibrio, Photobacterium, Grimontia, and Enterovibrio species. Growth pattern analysis demonstrated that V. coralliilyticus utilized myo-inositol as a sole carbon source, with a short lag time of 3 h. An iolG deletion mutant, which encodes an inositol dehydrogenase, was unable to grow on myo-inositol. Within the iol clusters were an MFS-type (iolT1) and an ABC-type (iolXYZ) transporter and analyses showed that both transported myo-inositol. IolG and IolA phylogeny among Vibrionaceae species showed different evolutionary histories indicating multiple acquisition events. Outside of Vibrionaceae, IolG was most closely related to IolG from a small group of Aeromonas fish and human pathogens and Providencia species. However, IolG from hypervirulent A. hydrophila strains clustered with IolG from Enterobacter, and divergently from Pectobacterium, Brenneria, and Dickeya plant pathogens. The iol cluster was also present within Aliiroseovarius, Burkholderia, Endozoicomonas, Halomonas, Labrenzia, Marinomonas, Marinobacterium, Cobetia, Pantoea, and Pseudomonas, of which many species were associated with marine flora and fauna.IMPORTANCEHost associated bacteria such as Vibrio coralliilyticus encounter competition for nutrients and have evolved metabolic strategies to better compete for food. Emerging studies show that myo-inositol is exchanged in the coral-algae symbiosis, is likely involved in signaling, but is also an osmolyte in algae. The bacterial consumption of myo-inositol could contribute to a breakdown of the coral-algae symbiosis during thermal stress or disrupt the coral microbiome. Phylogenetic analyses showed that the evolutionary history of myo-inositol metabolism is complex, acquired multiple times in Vibrio, but acquired once in many bacterial plant pathogens. Further analysis also showed that a conserved iol cluster is prevalent among many marine species (commensals, mutualists, and pathogens) associated with marine flora and fauna, algae, sponges, corals, molluscs, crustaceans, and fish.
Collapse
Affiliation(s)
| | - Rachel M. Loughran
- Microbiology Graduate Program, University of Delaware, Newark, Delaware, USA
| | - Blake Ushijima
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Gary P. Richards
- U.S. Department of Agriculture, Agricultural Research Service, Dover, Delaware, USA
| | - E. Fidelma Boyd
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
17
|
Ollitrault G, Achebouche R, Dreux A, Murail S, Audouze K, Tromelin A, Taboureau O. Pred-O3, a web server to predict molecules, olfactory receptors and odor relationships. Nucleic Acids Res 2024; 52:W507-W512. [PMID: 38661190 PMCID: PMC11223793 DOI: 10.1093/nar/gkae305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024] Open
Abstract
The sense of smell is a biological process involving volatile molecules that interact with proteins called olfactory receptors to transmit a nervous message that allows the recognition of a perceived odor. However, the relationships between odorant molecules, olfactory receptors and odors (O3) are far from being well understood due to the combinatorial olfactory codes and large family of olfactory receptors. This is the reason why, based on 5802 odorant molecules and their annotations to 863 olfactory receptors (human) and 7029 odors and flavors annotations, a web server called Pred-O3 has been designed to provide insights into olfaction. Predictive models based on Artificial Intelligence have been developed allowing to suggest olfactory receptors and odors associated with a new molecule. In addition, based on the encoding of the odorant molecule's structure, physicochemical features related to odors and/or olfactory receptors are proposed. Finally, based on the structural models of the 98 olfactory receptors a systematic docking protocol can be applied and suggest if a molecule can bind or not to an olfactory receptor. Therefore, Pred-O3 is well suited to aid in the design of new odorant molecules and assist in fragrance research and sensory neuroscience. Pred-O3 is accessible at ' https://odor.rpbs.univ-paris-diderot.fr/'.
Collapse
Affiliation(s)
| | | | - Antoine Dreux
- Inserm U1133, CNRS UMR 8251, Université Paris Cité, Paris, France
| | - Samuel Murail
- Inserm U1133, CNRS UMR 8251, Université Paris Cité, Paris, France
| | | | - Anne Tromelin
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, F-21000 Dijon, France
| | | |
Collapse
|
18
|
Bugnon M, Röhrig UF, Goullieux M, Perez MS, Daina A, Michielin O, Zoete V. SwissDock 2024: major enhancements for small-molecule docking with Attracting Cavities and AutoDock Vina. Nucleic Acids Res 2024; 52:W324-W332. [PMID: 38686803 PMCID: PMC11223881 DOI: 10.1093/nar/gkae300] [Citation(s) in RCA: 80] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/19/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024] Open
Abstract
Drug discovery aims to identify potential therapeutic compounds capable of modulating the activity of specific biological targets. Molecular docking can efficiently support this process by predicting binding interactions between small molecules and macromolecular targets and potentially accelerating screening campaigns. SwissDock is a computational tool released in 2011 as part of the SwissDrugDesign project, providing a free web-based service for small-molecule docking after automatized preparation of ligands and targets. Here, we present the latest version of SwissDock, in which EADock DSS has been replaced by two state-of-the-art docking programs, i.e. Attracting Cavities and AutoDock Vina. AutoDock Vina provides faster docking predictions, while Attracting Cavities offers more accurate results. Ligands can be imported in various ways, including as files, SMILES notation or molecular sketches. Targets can be imported as PDB files or identified by their PDB ID. In addition, advanced search options are available both for ligands and targets, giving users automatized access to widely-used databases. The web interface has been completely redesigned for interactive submission and analysis of docking results. Moreover, we developed a user-friendly command-line access which, in addition to all options of the web site, also enables covalent ligand docking with Attracting Cavities. The new version of SwissDock is freely available at https://www.swissdock.ch/.
Collapse
Affiliation(s)
- Marine Bugnon
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Ute F Röhrig
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Mathilde Goullieux
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Marta A S Perez
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Antoine Daina
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Olivier Michielin
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital (HUG), CH-1205 Geneva, Switzerland
| | - Vincent Zoete
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
- Department of Oncology UNIL-CHUV, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
19
|
Kochnev Y, Ahmed M, Maldonado A, Durrant J. MolModa: accessible and secure molecular docking in a web browser. Nucleic Acids Res 2024; 52:W498-W506. [PMID: 38783339 PMCID: PMC11223821 DOI: 10.1093/nar/gkae406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/14/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Molecular docking advances early-stage drug discovery by predicting the geometries and affinities of small-molecule compounds bound to drug-target receptors, predictions that researchers can leverage in prioritizing drug candidates for experimental testing. Unfortunately, existing docking tools often suffer from poor usability, data security, and maintainability, limiting broader adoption. Additionally, the complexity of the docking process, which requires users to execute a series of specialized steps, often poses a substantial barrier for non-expert users. Here, we introduce MolModa, a secure, accessible environment where users can perform molecular docking entirely in their web browsers. We provide two case studies that illustrate how MolModa provides valuable biological insights. We further compare MolModa to other docking tools to highlight its strengths and limitations. MolModa is available free of charge for academic and commercial use, without login or registration, at https://durrantlab.com/molmoda.
Collapse
Affiliation(s)
- Yuri Kochnev
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mayar Ahmed
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alex M Maldonado
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
20
|
McAndrew H, Rigler J, Yeguvapalli S, Chitrala KN. Analysis of gene expression profiles to elucidate racial differences in African American and White patients with Triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596478. [PMID: 38853921 PMCID: PMC11160710 DOI: 10.1101/2024.05.29.596478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Triple-negative breast cancer (TNBC) is the second most diagnosed subtype of breast cancer. It is known to be the most aggressive one that lacks known targetable receptors. One of the concerns in TNBC is the disparities in its prevalence and tumor pathogenesis among women with non-Hispanic African American backgrounds. Despite extensive research, the genetic underpinnings that lead to these disparities remain elusive. The current study aims to provide initiative for further clinical research in the development of targeted therapy for TNBC. Gene expression profiles from African American (AA) and European American (EA) patients with TNBC were collected from Gene Expression Omnibus and performed differential gene expression (DEG)analysis. Candidate genes for a significant correlation between expression and survival rates for breast invasive carcinoma were analyzed using UALCAN. The DAVID annotation tool, Enrichr web server, KEGG database, and Gene Ontology (GO) database were used for functional enrichment analysis of target genes. The Network Analyst server was used to identify ligands with strong affinities, SeamDock server for molecular docking between the biomarkers/associated ligands and examined protein-protein interactions (PPI) from the STRING server. Data from public breast cancer cohorts was utilized to identify expression patterns associated with poor survival outcomes of AA patients with TNBC. Our results showed three genes of interest ( CCT3 , LSM2 , and MRPS16 ) and potential ligands for molecular docking. Molecular docking was performed for the ICG001 ligand to CCT3 (binding affinities of -9.3 kcal/mol and -8.9 kcal/mol) and other interacting proteins ( CDC20 and PPP2CA ) with high degrees of connectivity. The results determined molecular docking of ICG001 to the CDC20 protein resulted in the highest binding affinity. Our results demonstrated that CCT3 and its interacting partners could serve as potential biomarkers due to their association with the survival outcome of AA patients with TNBC and ICG 001 could be the therapeutic lead for these biomarkers.
Collapse
|
21
|
Maghembe RS, Magulye MAK, Eilu E, Sekyanzi S, Makaranga A, Mwesigwa S, Katagirya E. A sophisticated virulence repertoire and colistin resistance of Citrobacter freundii ST150 from a patient with sepsis admitted to ICU in a tertiary care hospital in Uganda, East Africa: Insight from genomic and molecular docking analyses. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 120:105591. [PMID: 38604286 PMCID: PMC11069293 DOI: 10.1016/j.meegid.2024.105591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Sepsis and multidrug resistance comprise a complex of factors attributable to mortality among intensive care unit (ICU) patients globally. Pathogens implicated in sepsis are diverse, and their virulence and drug resistance remain elusive. From a tertiary care hospital ICU in Uganda, we isolated a Citrobacter freundii strain RSM030 from a patient with sepsis and phenotypically tested it against a panel of 16 antibiotics including imipenem levofloxacin, cotrimoxazole and colistin, among others. We sequenced the organism's genome and integrated multilocus sequencing (MLST), PathogenFinder with Virulence Factor analyzer (VFanalyzer) to establish its pathogenic relevance. Thereafter, we combined antiSMASH and PRISM genome mining with molecular docking to predict biosynthetic gene clusters (BGCs), pathways, toxin structures and their potential targets in-silico. Finally, we coupled ResFinder with comprehensive antibiotic resistance database (CARD) to scrutinize the genomic antimicrobial resistance profile of the isolate. From PathogenFinder and MLST, this organism was confirmed to be a human pathogen (p = 0.843), sequence type (ST)150, whose virulence is determined by chromosomal type III secretion system (T3SS) (the injectosome) and plasmid-encoded type IV secretion system (T4SS), the enterobactin biosynthetic gene cluster and biofilm formation through the pgaABCD operon. Pathway and molecular docking analyses revealed that the shikimate pathway can generate a toxin targeting multiple host proteins including spectrin, detector of cytokinesis protein 2 (Dock2) and plasmalemma vesicle-associated protein (PLVAP), potentially distorting the host cell integrity. From phenotypic antibiotic testing, we found indeterminate results for amoxicillin/clavulanate and levofloxacin, with resistance to cotrimoxazole and colistin. Detailed genome analysis revealed chromosomal beta lactam resistance genes, i.e. blaCMY-79, blaCMY-116 and blaTEM-1B, along with multiple mutations of the lipopolysaccharide modifying operon genes PmrA/PmrB, pmrD, mgrA/mgrB and PhoP/PhoQ, conferring colistin resistance. From these findings, we infer that Citrobacter freundii strain RSM030 is implicated in sepsis and resistance to standard antibiotics, including colistin, the last resort.
Collapse
Affiliation(s)
- Reuben S Maghembe
- Biological and Marine Sciences Unit, Faculty of Natural and Applied Sciences, Marian University College, P. O. Box 47, Bagamoyo, Tanzania; Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda; Department of Microbiology and Immunology, Faculty of Biomedical Sciences, Kampala International University, Wester Campus, Ishaka, Uganda.; Microbiology Section, Department of Biological Sciences, University of Botswana, Private Bag 0704, Gaborone, Botswana.
| | - Maximilian A K Magulye
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Emmanuel Eilu
- Department of Microbiology and Immunology, Faculty of Biomedical Sciences, Kampala International University, Wester Campus, Ishaka, Uganda
| | - Simon Sekyanzi
- Department of Medical Microbiology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Abdalah Makaranga
- Biological and Marine Sciences Unit, Faculty of Natural and Applied Sciences, Marian University College, P. O. Box 47, Bagamoyo, Tanzania
| | - Savannah Mwesigwa
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Eric Katagirya
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| |
Collapse
|
22
|
Mistretta M, Cimino M, Campagne P, Volant S, Kornobis E, Hebert O, Rochais C, Dallemagne P, Lecoutey C, Tisnerat C, Lepailleur A, Ayotte Y, LaPlante SR, Gangneux N, Záhorszká M, Korduláková J, Vichier-Guerre S, Bonhomme F, Pokorny L, Albert M, Tinevez JY, Manina G. Dynamic microfluidic single-cell screening identifies pheno-tuning compounds to potentiate tuberculosis therapy. Nat Commun 2024; 15:4175. [PMID: 38755132 PMCID: PMC11099131 DOI: 10.1038/s41467-024-48269-2] [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: 03/13/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Drug-recalcitrant infections are a leading global-health concern. Bacterial cells benefit from phenotypic variation, which can suggest effective antimicrobial strategies. However, probing phenotypic variation entails spatiotemporal analysis of individual cells that is technically challenging, and hard to integrate into drug discovery. In this work, we develop a multi-condition microfluidic platform suitable for imaging two-dimensional growth of bacterial cells during transitions between separate environmental conditions. With this platform, we implement a dynamic single-cell screening for pheno-tuning compounds, which induce a phenotypic change and decrease cell-to-cell variation, aiming to undermine the entire bacterial population and make it more vulnerable to other drugs. We apply this strategy to mycobacteria, as tuberculosis poses a major public-health threat. Our lead compound impairs Mycobacterium tuberculosis via a peculiar mode of action and enhances other anti-tubercular drugs. This work proves that harnessing phenotypic variation represents a successful approach to tackle pathogens that are increasingly difficult to treat.
Collapse
Affiliation(s)
- Maxime Mistretta
- Institut Pasteur, Université Paris Cité, Microbial Individuality and Infection Laboratory, 75015, Paris, France
| | - Mena Cimino
- Institut Pasteur, Université Paris Cité, Microbial Individuality and Infection Laboratory, 75015, Paris, France
| | - Pascal Campagne
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015, Paris, France
| | - Stevenn Volant
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015, Paris, France
| | - Etienne Kornobis
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015, Paris, France
- Institut Pasteur, Université Paris Cité, Biomics Platform, 75015, Paris, France
| | | | | | | | | | | | | | - Yann Ayotte
- Institut National de la Recherche Scientifique-Armand-Frappier Santé Biotechnologie Research Centre, Laval, Quebec, H7V 1B7, Canada
| | - Steven R LaPlante
- Institut National de la Recherche Scientifique-Armand-Frappier Santé Biotechnologie Research Centre, Laval, Quebec, H7V 1B7, Canada
| | - Nicolas Gangneux
- Institut Pasteur, Université Paris Cité, Microbial Individuality and Infection Laboratory, 75015, Paris, France
| | - Monika Záhorszká
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15, Bratislava, Slovakia
| | - Jana Korduláková
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15, Bratislava, Slovakia
| | - Sophie Vichier-Guerre
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Epigenetic Chemical Biology Unit, 75015, Paris, France
| | - Frédéric Bonhomme
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Epigenetic Chemical Biology Unit, 75015, Paris, France
| | - Laura Pokorny
- Institut Pasteur, Université Paris Cité, Microbial Individuality and Infection Laboratory, 75015, Paris, France
| | - Marvin Albert
- Institut Pasteur, Université Paris Cité, Image Analysis Hub, 75015, Paris, France
| | - Jean-Yves Tinevez
- Institut Pasteur, Université Paris Cité, Image Analysis Hub, 75015, Paris, France
| | - Giulia Manina
- Institut Pasteur, Université Paris Cité, Microbial Individuality and Infection Laboratory, 75015, Paris, France.
| |
Collapse
|
23
|
Maghembe RS, Magulye MA, Eilu E, Sekyanzi S, Mwesigwa S, Katagirya E. Chromosomal and plasmid-encoded virulence and multidrug resistance of Escherichia coli ST58/24 infecting a 2-year-old sickle cell patient with sepsis in Kampala Uganda, East Africa. Heliyon 2024; 10:e30187. [PMID: 38707307 PMCID: PMC11068601 DOI: 10.1016/j.heliyon.2024.e30187] [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: 01/26/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
Sepsis and drug resistance represent a complex of the most common global causes of mortality in intensive care units (ICUs) especially among patients with comorbidities. Extraintestinal pathogenic Escherichia coli (ExPEC) strains are highly implicated in systemic infections, with multidrug resistance exacerbating the risk of chronic conditions and patient mortality. The diversity of virulence and evolution of multidrug resistance are yet to be fully deciphered. In this work, we aimed at unveiling the pathogens and their genomic determinants of virulence and drug resistance relevant to increased sepsis in a sickle cell child admitted to ICU. From a rectal swab, we isolated a strain of E. coli from the patient and phenotypically tested it against a panel of selected beta lactams, fluoroquinolones, macrolides, aminoglycosides and colistin. We then sequenced the entire genome and integrated multiple bioinformatic pipelines to divulge the virulence and multidrug resistance profiles of the isolate. Our results revealed that the isolate belongs to the sequence type (ST) 58/24, which (ST58), is a known ExPEC. With the use of PathogenFinder, we were able to confirm that this isolate is a human pathogen (p = 0.936). The assembled chromosome and two plasmids encode virulence factors related to capsule (antiphagocytosis), serum survival and resistance, type 6 secretion system (T6SS), multiple siderophores (iron acquisition), and biosynthetic gene clusters for polyketides and nonribosomal peptides exhibiting host cell damaging activity in silico. The genome also harbors multidrug resistance genotypes including extended spectrum beta lactamase (ESBL) genes such as blaTEM-1A/B, sulfonamide resistance genes sul1/2, fluoroquinolone resistance genes dfrA5 and nonsynonymous mutations of the gene pmrB, conferring intrinsic colistin resistance. Conclusively, this pathogen holds the potential to cause systemic infection and might exacerbate sickle cell anemia in the patient. The virulence and multidrug resistance profiles are encoded by both the chromosome and plasmids. Genomic surveillance of pathogens with multidrug resistance among patients with commodities is crucial for effective disease management.
Collapse
Affiliation(s)
- Reuben S. Maghembe
- Department of Microbiologgy and Immunology, Faculty of Biomedical Sciences, Kampala International University, Western Campus, Ishaka, Uganda
- Department of Health and Biomedical Sciences, Didia Education and Health Organization, P. O. Box 113, Shinyanga, Tanzania
- Biological and Marine Sciences Unit, Faculty of Natural and Applied Sciences, Marian University College, P. O. Box 47, Bagamoyo, Tanzania
- Microbiology Section, Department of Biological Sciences, Faculty of Science, University of Botswana, Private Bag 00704, Gaborone, Botswana
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Maximilian A.K. Magulye
- Department of Health and Biomedical Sciences, Didia Education and Health Organization, P. O. Box 113, Shinyanga, Tanzania
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Emmanuel Eilu
- Department of Microbiologgy and Immunology, Faculty of Biomedical Sciences, Kampala International University, Western Campus, Ishaka, Uganda
| | - Simon Sekyanzi
- Department of Medical Microbiology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Savannah Mwesigwa
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Eric Katagirya
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| |
Collapse
|
24
|
Boas Lichty KE, Loughran RM, Ushijima B, Richards GP, Boyd EF. Osmotic stress response of the coral and oyster pathogen Vibrio coralliilyticus : acquisition of catabolism gene clusters for the compatible solute and signaling molecule myo -inositol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575920. [PMID: 38766061 PMCID: PMC11100586 DOI: 10.1101/2024.01.16.575920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Marine bacteria experience fluctuations in osmolarity that they must adapt to, and most bacteria respond to high osmolarity by accumulating compatible solutes also known as osmolytes. The osmotic stress response and compatible solutes used by the coral and oyster pathogen Vibrio coralliilyticus were unknown. In this study, we showed that to alleviate osmotic stress V. coralliilyticus biosynthesized glycine betaine (GB) and transported into the cell choline, GB, ectoine, dimethylglycine, and dimethylsulfoniopropionate, but not myo -inositol. Myo -inositol is a stress protectant and a signaling molecule that is biosynthesized and used by algae. Bioinformatics identified myo -inositol ( iol ) catabolism clusters in V. coralliilyticus and other Vibrio, Photobacterium, Grimontia, and Enterovibrio species. Growth pattern analysis demonstrated that V. coralliilyticus utilized myo -inositol as a sole carbon source, with a short lag time of 3 h. An iolG deletion mutant, which encodes an inositol dehydrogenase, was unable to grow on myo -inositol. Within the iol clusters were an MFS-type ( iolT1) and an ABC-type ( iolXYZ) transporter and analyses showed that both transported myo -inositol. IolG and IolA phylogeny among Vibrionaceae species showed different evolutionary histories indicating multiple acquisition events. Outside of Vibrionaceae , IolG was most closely related to IolG from a small group of Aeromonas fish and human pathogens and Providencia species. However, IolG from hypervirulent A. hydrophila strains clustered with IolG from Enterobacter, and divergently from Pectobacterium, Brenneria, and Dickeya plant pathogens. The iol cluster was also present within Aliiroseovarius, Burkholderia, Endozoicomonas, Halomonas, Labrenzia, Marinomonas, Marinobacterium, Cobetia, Pantoea, and Pseudomonas, of which many species were associated with marine flora and fauna. IMPORTANCE Host associated bacteria such as V. coralliilyticus encounter competition for nutrients and have evolved metabolic strategies to better compete for food. Emerging studies show that myo -inositol is exchanged in the coral-algae symbiosis, is likely involved in signaling, but is also an osmolyte in algae. The bacterial consumption of myo -inositol could contribute to a breakdown of the coral-algae symbiosis during thermal stress or disrupt the coral microbiome. Phylogenetic analyses showed that the evolutionary history of myo -inositol metabolism is complex, acquired multiple times in Vibrio, but acquired once in many bacterial plant pathogens. Further analysis also showed that a conserved iol cluster is prevalent among many marine species (commensals, mutualists, and pathogens) associated with marine flora and fauna, algae, sponges, corals, molluscs, crustaceans, and fish.
Collapse
|
25
|
Tang J, Matsuda Y. Functional analysis of transmembrane terpene cyclases involved in fungal meroterpenoid biosynthesis. Methods Enzymol 2024; 699:419-445. [PMID: 38942513 DOI: 10.1016/bs.mie.2024.02.007] [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: 06/30/2024]
Abstract
Pyr4-family terpene cyclases are noncanonical transmembrane class II terpene cyclases that catalyze a variety of cyclization reactions in the biosynthesis of microbial terpenoids, such as meroterpenoids. However, although these cyclases are widely distributed in microorganisms, their three-dimensional structures have not been determined, possibly due to the transmembrane locations of these enzymes. In this chapter, we describe procedures for the functional analysis of transmembrane terpene cyclases based on their model structures generated using AlphaFold2. We used AdrI, the Pyr4-family terpene cyclase required for the biosynthesis of andrastin A and its homologs, as an example.
Collapse
Affiliation(s)
- Jia Tang
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, P.R. China
| | - Yudai Matsuda
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR, P.R. China.
| |
Collapse
|
26
|
Knight IS, Mailhot O, Tang KG, Irwin JJ. DockOpt: A Tool for Automatic Optimization of Docking Models. J Chem Inf Model 2024; 64:1004-1016. [PMID: 38206771 PMCID: PMC10865354 DOI: 10.1021/acs.jcim.3c01406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/17/2023] [Accepted: 12/26/2023] [Indexed: 01/13/2024]
Abstract
Molecular docking is a widely used technique for leveraging protein structure for ligand discovery, but it remains difficult to utilize due to limitations that have not been adequately addressed. Despite some progress toward automation, docking still requires expert guidance, hindering its adoption by a broader range of investigators. To make docking more accessible, we developed a new utility called DockOpt, which automates the creation, evaluation, and optimization of docking models prior to their deployment in large-scale prospective screens. DockOpt outperforms our previous automated pipeline across all 43 targets in the DUDE-Z benchmark data set, and the generated models for 84% of targets demonstrate sufficient enrichment to warrant their use in prospective screens, with normalized LogAUC values of at least 15%. DockOpt is available as part of the Python package Pydock3 included in the UCSF DOCK 3.8 distribution, which is available for free to academic researchers at https://dock.compbio.ucsf.edu and free for everyone upon registration at https://tldr.docking.org.
Collapse
Affiliation(s)
- Ian S. Knight
- Department of Pharmaceutical Chemistry, UCSF, 1700 Fourth Street, San Francisco, California 94158-2330, United States
| | - Olivier Mailhot
- Department of Pharmaceutical Chemistry, UCSF, 1700 Fourth Street, San Francisco, California 94158-2330, United States
| | - Khanh G. Tang
- Department of Pharmaceutical Chemistry, UCSF, 1700 Fourth Street, San Francisco, California 94158-2330, United States
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, UCSF, 1700 Fourth Street, San Francisco, California 94158-2330, United States
| |
Collapse
|
27
|
Buntin K, Mrak P, Pivk Lukančič P, Wollbrett S, Drčar T, Krastel P, Thibaut C, Salcius M, Gao X, Wang S, Weber E, Koplan E, Regenass H. Generation of Bioactivity-Tailored FK506/FK520 Analogs by CRISPR Editing in Streptomyces tsukubaensis. Chemistry 2024; 30:e202302350. [PMID: 37855054 DOI: 10.1002/chem.202302350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
For a potential application of FK506 in the treatment of acute kidney failure only the FKBP12 binding capability of the compound is required, while the immunosuppressive activity via calcineurin binding is considered as a likely risk to the patients. The methoxy groups at C13 and C15 are thought to have significant influence on the immunosuppressive activity of the molecule. Consequently, FK506 analogs with different functionalities at C13 and C15 were generated by targeted CRISPR editing of the AT domains in module 7 and 8 of the biosynthetic assembly line in Streptomyces tsukubaensis. In addition, the corresponding FK520 (C21 ethyl derivative of FK506) analogs could be obtained by media adjustments. The compounds were tested for their bioactivity in regards to FKBP12 binding, BMP potentiation and calcineurin sparing. 15-desmethoxy FK506 was superior to the other tested analogs as it did not inhibit calcineurin but retained high potency towards FKBP12 binding and BMP potentiation.
Collapse
Affiliation(s)
- Kathrin Buntin
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Peter Mrak
- Manufacturing Scienes & Technologies, Sandoz Technical Operations, Lek Pharmaceuticals d.d., Kolodvorska 27, 1234, Mengeš, Slovenia
| | - Petra Pivk Lukančič
- Manufacturing Scienes & Technologies, Sandoz Technical Operations, Lek Pharmaceuticals d.d., Kolodvorska 27, 1234, Mengeš, Slovenia
| | - Séverine Wollbrett
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Tjasa Drčar
- Manufacturing Scienes & Technologies, Sandoz Technical Operations, Lek Pharmaceuticals d.d., Kolodvorska 27, 1234, Mengeš, Slovenia
| | - Philipp Krastel
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Christian Thibaut
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Michael Salcius
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Inc. 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Xiaolin Gao
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Inc. 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Shaowen Wang
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Inc. 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Eric Weber
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Eva Koplan
- Manufacturing Scienes & Technologies, Sandoz Technical Operations, Lek Pharmaceuticals d.d., Kolodvorska 27, 1234, Mengeš, Slovenia
| | - Hugo Regenass
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| |
Collapse
|
28
|
Flachsenberg F, Ehrt C, Gutermuth T, Rarey M. Redocking the PDB. J Chem Inf Model 2024; 64:219-237. [PMID: 38108627 DOI: 10.1021/acs.jcim.3c01573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Molecular docking is a standard technique in structure-based drug design (SBDD). It aims to predict the 3D structure of a small molecule in the binding site of a receptor (often a protein). Despite being a common technique, it often necessitates multiple tools and involves manual steps. Here, we present the JAMDA preprocessing and docking workflow that is easy to use and allows fully automated docking. We evaluate the JAMDA docking workflow on binding sites extracted from the complete PDB and derive key factors determining JAMDA's docking performance. With that, we try to remove most of the bias due to manual intervention and provide a realistic estimate of the redocking performance of our JAMDA preprocessing and docking workflow for any PDB structure. On this large PDBScan22 data set, our JAMDA workflow finds a pose with an RMSD of at most 2 Å to the crystal ligand on the top rank for 30.1% of the structures. When applying objective structure quality filters to the PDBScan22 data set, the success rate increases to 61.8%. Given the prepared structures from the JAMDA preprocessing pipeline, both JAMDA and the widely used AutoDock Vina perform comparably on this filtered data set (the PDBScan22-HQ data set).
Collapse
Affiliation(s)
- Florian Flachsenberg
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - Christiane Ehrt
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - Torben Gutermuth
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - Matthias Rarey
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| |
Collapse
|
29
|
Fayez AG, Esmaiel NN, Ashaat EA, Refeat MM, Lotfy RS, Raouf HA, El Ruby MO. New drug-like small molecule antagonizes phosphatidylinositol (3,4,5)-trisphosphate (PIP3) in patients with conotruncal heart defects. J Taibah Univ Med Sci 2023; 18:1244-1253. [PMID: 37250809 PMCID: PMC10213100 DOI: 10.1016/j.jtumed.2023.04.006] [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: 09/18/2022] [Revised: 02/02/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Objectives Conotruncal heart defects (CTDs) are highly heritable, and approximately one-third of all congenital heart defects are due to CTDs. Through post-analysis of GWAS data relevant to CTDs, a new putative signal transduction pathway, called Vars2-Pic3ca-Akt, associated with CTD has been hypothesized. Here, we aimed to validate the Vars2-Pic3ca-Akt pathway experimentally by measuring Vars2 and PIP3 in patients with CTDs and controls, and to construct a PIP3 inhibitor, as one of harmful-relevant CTD pathogenesis, through an Akt-based drug design strategy. Methods rs2517582 genotype and relative Vars2 expression in 207 individuals were determined by DNA sequencing and qPCR respectively, and free plasma PIP3 in 190 individuals was quantified through ELISA. An Akt-pharmacophore feature model was used to discover PIP3 antagonists with multiple computational and drug-like estimation tools. Results CTD pathogenesis due to Vars2-Pic3ca-Akt overstimulation was confirmed by elevated Vars2 and PIP3 in patients with CTDs. We identified a new small molecule, 322PESB, that antagonizes PIP3 binding. This molecule was prioritized via virtual screening of 21 hypothetical small molecules and it showed minimal RMSD change, high binding affinity andlower dissociation constant than PIP3-Akt complex by 1.99 Kcal/Mol, thus resulting in an equilibrium shift toward 322PESB-Akt complex formation. Moreover, 322PESB exhibited acceptable pharmacokinetics and drug likeness features according to ADME and Lipinski's rule of five classifiers. This compound is the first potential drug-like molecule reported for patients with CTDs with elevated PIP3. Conclusion PIP3 is a useful diagnostic biomarker for patients with CTDs. The Akt-pharmacophore feature model is a feasible approach for discovery of PIP3 signalling antagonists. Further 322PESB development and testing are recommended.
Collapse
Affiliation(s)
- Alaaeldin G. Fayez
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Nora N. Esmaiel
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Engy A. Ashaat
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Miral M. Refeat
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Randa S. Lotfy
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Haiam Abdel Raouf
- Immunogenetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| | - Mona O. El Ruby
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Giza, Egypt
| |
Collapse
|
30
|
Krause F, Voigt K, Di Ventura B, Öztürk MA. ReverseDock: a web server for blind docking of a single ligand to multiple protein targets using AutoDock Vina. Front Mol Biosci 2023; 10:1243970. [PMID: 37881441 PMCID: PMC10594994 DOI: 10.3389/fmolb.2023.1243970] [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: 06/21/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023] Open
Abstract
Several platforms exist to perform molecular docking to computationally predict binders to a specific protein target from a library of ligands. The reverse, that is, docking a single ligand to various protein targets, can currently be done by very few web servers, which limits the search to a small set of pre-selected human proteins. However, the possibility to in silico predict which targets a compound identified in a high-throughput drug screen bind would help optimize and reduce the costs of the experimental workflow needed to reveal the molecular mechanism of action of a ligand. Here, we present ReverseDock, a blind docking web server based on AutoDock Vina specifically designed to allow users with no computational expertise to dock a ligand to 100 protein structures of their choice. ReverseDock increases the number and type of proteins a ligand can be docked to, making the task of in silico docking of a ligand to entire families of proteins straightforward. We envision ReverseDock will support researchers by providing the possibility to apply inverse docking computations using web browser. ReverseDock is available at: https://reversedock.biologie.uni-freiburg.de/.
Collapse
Affiliation(s)
- Fabian Krause
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Biology II, University of Freiburg, Freiburg, Germany
| | - Karsten Voigt
- Institute of Biology III, University of Freiburg, Freiburg, Germany
| | - Barbara Di Ventura
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Biology II, University of Freiburg, Freiburg, Germany
| | - Mehmet Ali Öztürk
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Biology II, University of Freiburg, Freiburg, Germany
| |
Collapse
|
31
|
Simionescu D, Tharayil N, Leonard E, Carlyle W, Schwarz A, Ning K, Carsten C, Garcia JCC, Carter A, Owens C, Simionescu A. Binding of Pentagalloyl Glucose to Aortic Wall Proteins: Insights from Peptide Mapping and Simulated Docking Studies. Bioengineering (Basel) 2023; 10:936. [PMID: 37627822 PMCID: PMC10451288 DOI: 10.3390/bioengineering10080936] [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: 06/12/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Pentagalloyl glucose (PGG) is currently being investigated as a non-surgical treatment for abdominal aortic aneurysms (AAAs); however, the molecular mechanisms of action of PGG on the AAA matrix components and the intra-luminal thrombus (ILT) still need to be better understood. To assess these interactions, we utilized peptide fingerprinting and molecular docking simulations to predict the binding of PGG to vascular proteins in normal and aneurysmal aorta, including matrix metalloproteinases (MMPs), cytokines, and fibrin. We performed PGG diffusion studies in pure fibrin gels and human ILT samples. PGG was predicted to bind with high affinity to most vascular proteins, the active sites of MMPs, and several cytokines known to be present in AAAs. Finally, despite potential binding to fibrin, PGG was shown to diffuse readily through thrombus at physiologic pressures. In conclusion, PGG can bind to all the normal and aneurysmal aorta protein components with high affinity, potentially protecting the tissue from degradation and exerting anti-inflammatory activities. Diffusion studies showed that thrombus presence in AAAs is not a barrier to endovascular treatment. Together, these results provide a deeper understanding of the clinical potential of PGG as a non-surgical treatment of AAAs.
Collapse
Affiliation(s)
- Dan Simionescu
- Biocompatibility and Tissue Regeneration Laboratory, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; (J.C.C.G.); (A.C.)
| | - Nishanth Tharayil
- Multi-User Analytical Lab (MUAL) & Metabolomic Core, Clemson University, Clemson, SC 29634, USA; (N.T.); (E.L.)
| | - Elizabeth Leonard
- Multi-User Analytical Lab (MUAL) & Metabolomic Core, Clemson University, Clemson, SC 29634, USA; (N.T.); (E.L.)
| | - Wenda Carlyle
- Nectero Medical Inc., Mesa, AZ 85281, USA; (W.C.); (A.S.); (K.N.)
| | - Alex Schwarz
- Nectero Medical Inc., Mesa, AZ 85281, USA; (W.C.); (A.S.); (K.N.)
| | - Kelvin Ning
- Nectero Medical Inc., Mesa, AZ 85281, USA; (W.C.); (A.S.); (K.N.)
| | | | - Juan Carlos Carrillo Garcia
- Biocompatibility and Tissue Regeneration Laboratory, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; (J.C.C.G.); (A.C.)
| | - Alexander Carter
- Biocompatibility and Tissue Regeneration Laboratory, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; (J.C.C.G.); (A.C.)
| | - Collin Owens
- Tissue Engineering Laboratory, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; (C.O.); (A.S.)
| | - Agneta Simionescu
- Tissue Engineering Laboratory, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; (C.O.); (A.S.)
| |
Collapse
|
32
|
Masum MHU, Rajia S, Bristi UP, Akter MS, Amin MR, Shishir TA, Ferdous J, Ahmed F, Rahaman MM, Saha O. In Silico Functional Characterization of a Hypothetical Protein From Pasteurella Multocida Reveals a Novel S-Adenosylmethionine-Dependent Methyltransferase Activity. Bioinform Biol Insights 2023; 17:11779322231184024. [PMID: 37424709 PMCID: PMC10328030 DOI: 10.1177/11779322231184024] [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: 04/01/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Genomes may now be sequenced in a matter of weeks, leading to an influx of "hypothetical" proteins (HP) whose activities remain a mystery in GenBank. The information included inside these genes has quickly grown in prominence. Thus, we selected to look closely at the structure and function of an HP (AFF25514.1; 246 residues) from Pasteurella multocida (PM) subsp. multocida str. HN06. Possible insights into bacterial adaptation to new environments and metabolic changes might be gained by studying the functions of this protein. The PM HN06 2293 gene encodes an alkaline cytoplasmic protein with a molecular weight of 28352.60 Da, an isoelectric point (pI) of 9.18, and an overall average hydropathicity of around -0.565. One of its functional domains, tRNA (adenine (37)-N6)-methyltransferase TrmO, is a S-adenosylmethionine (SAM)-dependent methyltransferase (MTase), suggesting that it belongs to the Class VIII SAM-dependent MTase family. The tertiary structures represented by HHpred and I-TASSER models were found to be flawless. We predicted the model's active site using the Computed Atlas of Surface Topography of Proteins (CASTp) and FTSite servers, and then displayed it in 3 dimensional (3D) using PyMOL and BIOVIA Discovery Studio. Based on molecular docking (MD) results, we know that HP interacts with SAM and S-adenosylhomocysteine (SAH), 2 crucial metabolites in the tRNA methylation process, with binding affinities of 7.4 and 7.5 kcal/mol, respectively. Molecular dynamic simulations (MDS) of the docked complex, which included only modest structural adjustments, corroborated the strong binding affinity of SAM and SAH to the HP. Evidence for HP's possible role as an SAM-dependent MTase was therefore given by the findings of Multiple sequence alignment (MSA), MD, and molecular dynamic modeling. These in silico data suggest that the investigated HP might be used as a useful adjunct in the investigation of Pasteurella infections and the development of drugs to treat zoonotic pasteurellosis.
Collapse
Affiliation(s)
- Md. Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Sultana Rajia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Uditi Paul Bristi
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Mir Salma Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Mohammad Ruhul Amin
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Tushar Ahmed Shishir
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Department of Medicine, Abdul Malek Ukil Medical College, Noakhali, Bangladesh
| | - Firoz Ahmed
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Otun Saha
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
33
|
Vistoli G, Manelfi C, Talarico C, Fava A, Warshel A, Tetko IV, Apostolov R, Ye Y, Latini C, Ficarelli F, Palermo G, Gadioli D, Vitali E, Varriale G, Pisapia V, Scaturro M, Coletti S, Gregori D, Gruffat D, Leija E, Hessenauer S, Delbianco A, Allegretti M, Beccari AR. MEDIATE - Molecular DockIng at homE: Turning collaborative simulations into therapeutic solutions. Expert Opin Drug Discov 2023; 18:821-833. [PMID: 37424369 DOI: 10.1080/17460441.2023.2221025] [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: 11/18/2022] [Accepted: 05/30/2023] [Indexed: 07/11/2023]
Abstract
INTRODUCTION Collaborative computing has attracted great interest in the possibility of joining the efforts of researchers worldwide. Its relevance has further increased during the pandemic crisis since it allows for the strengthening of scientific collaborations while avoiding physical interactions. Thus, the E4C consortium presents the MEDIATE initiative which invited researchers to contribute via their virtual screening simulations that will be combined with AI-based consensus approaches to provide robust and method-independent predictions. The best compounds will be tested, and the biological results will be shared with the scientific community. AREAS COVERED In this paper, the MEDIATE initiative is described. This shares compounds' libraries and protein structures prepared to perform standardized virtual screenings. Preliminary analyses are also reported which provide encouraging results emphasizing the MEDIATE initiative's capacity to identify active compounds. EXPERT OPINION Structure-based virtual screening is well-suited for collaborative projects provided that the participating researchers work on the same input file. Until now, such a strategy was rarely pursued and most initiatives in the field were organized as challenges. The MEDIATE platform is focused on SARS-CoV-2 targets but can be seen as a prototype which can be utilized to perform collaborative virtual screening campaigns in any therapeutic field by sharing the appropriate input files.
Collapse
Affiliation(s)
- Giulio Vistoli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | | | | | - Anna Fava
- EXSCALATE, Dompé Farmaceutici S.p.A, Napoli, Italy
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, USA
| | - Igor V Tetko
- BIGCHEM GmbH, Valerystr, Germany
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich-Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Rossen Apostolov
- PDC Center For High Performance Computing, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yang Ye
- Natural Products Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chiara Latini
- High Performance Computing Dept, CINECA, Casalecchio di Reno, Bologna, Italy
| | - Federico Ficarelli
- High Performance Computing Dept, CINECA, Casalecchio di Reno, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Rouzbeh N, Rau AR, Benton AJ, Yi F, Anderson CM, Johns MR, Jensen L, Lotti JS, Holley DC, Hansen KB. Allosteric modulation of GluN1/GluN3 NMDA receptors by GluN1-selective competitive antagonists. J Gen Physiol 2023; 155:e202313340. [PMID: 37078900 PMCID: PMC10125900 DOI: 10.1085/jgp.202313340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
NMDA-type ionotropic glutamate receptors are critical for normal brain function and are implicated in central nervous system disorders. Structure and function of NMDA receptors composed of GluN1 and GluN3 subunits are less understood compared to those composed of GluN1 and GluN2 subunits. GluN1/3 receptors display unusual activation properties in which binding of glycine to GluN1 elicits strong desensitization, while glycine binding to GluN3 alone is sufficient for activation. Here, we explore mechanisms by which GluN1-selective competitive antagonists, CGP-78608 and L-689,560, potentiate GluN1/3A and GluN1/3B receptors by preventing glycine binding to GluN1. We show that both CGP-78608 and L-689,560 prevent desensitization of GluN1/3 receptors, but CGP-78608-bound receptors display higher glycine potency and efficacy at GluN3 subunits compared to L-689,560-bound receptors. Furthermore, we demonstrate that L-689,560 is a potent antagonist of GluN1FA+TL/3A receptors, which are mutated to abolish glycine binding to GluN1, and that this inhibition is mediated by a non-competitive mechanism involving binding to the mutated GluN1 agonist binding domain (ABD) to negatively modulate glycine potency at GluN3A. Molecular dynamics simulations reveal that CGP-78608 and L-689,560 binding or mutations in the GluN1 glycine binding site promote distinct conformations of the GluN1 ABD, suggesting that the GluN1 ABD conformation influences agonist potency and efficacy at GluN3 subunits. These results uncover the mechanism that enables activation of native GluN1/3A receptors by application of glycine in the presence of CGP-78608, but not L-689,560, and demonstrate strong intra-subunit allosteric interactions in GluN1/3 receptors that may be relevant to neuronal signaling in brain function and disease.
Collapse
Affiliation(s)
- Nirvan Rouzbeh
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Andrew R. Rau
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Avery J. Benton
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - Feng Yi
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Carly M. Anderson
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Mia R. Johns
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Loren Jensen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - James S. Lotti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - David C. Holley
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - Kasper B. Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| |
Collapse
|
35
|
Dawood AA. The efficacy of Paxlovid against COVID-19 is the result of the tight molecular docking between M pro and antiviral drugs (nirmatrelvir and ritonavir). Adv Med Sci 2023; 68:1-9. [PMID: 36368287 PMCID: PMC9626444 DOI: 10.1016/j.advms.2022.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/05/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022]
Abstract
Purpose Currently, a number of medications for coronavirus disease 2019 (COVID-19) treatment are tested in clinical trials; however, credible clinical studies are becoming increasingly difficult to come by. Paxlovid is a ritonavir-boosted nirmatrelvir drug that the U.S. Food and Drug Administration (FDA) authorized for the treatment of COVID-19. This study aimed to demonstrate the interaction of nirmatrelvir and ritonavir on the active site of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro). Materials and methods To locate the optimal docking between Mpro and antiviral drugs, and to conduct dynamic simulations between atoms in the fusion areas, various bioinformatics and mathematical equations were applied. Results According to the docking data, nirmatrelvir has a stronger interaction with Mpro than ritonavir, which has more multiple bonds. Molecular docking of antiviral drugs such as Paxlovid has a significant impact on the treatment of COVID-19 virus. Conclusions According to this study, Paxlovid may work on new strains, including Omicron, because the Mpro mutation P132H in the Omicron variant has no direct effect on the protein.
Collapse
|
36
|
Varun E, Bhakti K, Aishwarya K, Suraj RH, Jagadish MR, Mohana Kumara P. Rohitukine content across the geographical distribution of Dysoxylum binectariferum Hook F. and its natural derivatives as potential sources of CDK inhibitors. Heliyon 2023; 9:e13469. [PMID: 36852056 PMCID: PMC9958448 DOI: 10.1016/j.heliyon.2023.e13469] [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: 09/02/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Dysoxylum binectariferum is an important medicinal plant distributed in the Western Ghats of India. The species has gained international importance for its anticancer component, rohitukine, a chromone alkaloid. Flavopiridol, P-276-00 and IIIM-290 are the derivatives of rohitukine in clinical trials against a wide range of cancers. Flavopiridol was recently approved as an orphan drug for chronic lymphocytic leukemia treatment. In this study, we report the isolation and characterization of rohitukine from the bark of D. binectariferum. Further, rohitukine was estimated across the Western-Ghats and the North-East regions of India. Additionally, D. binectariferum is also reported (∼45 compounds) to produce many natural derivatives of rohitukine and terpenoids, which were investigated in-silico to reveal promising CDK inhibitors. The metabolite fingerprinting of tissues of D. binectariferum was studied using HPTLC and FTIR. The distribution of major chromone alkaloid rohitukine was estimated by HPLC. Further, the pharmacological potential of D. binectariferum compounds was evaluated in-silico by discovering the potential protein targets, molecular docking, ADMET analysis and MD simulation. The isolation of rohitukine has yielded 0.6% from the bark of D. binectariferum. A higher percent of rohitukine was found in the Jog populations (0.58% & 1.28%: leaf & bark), whereas least was observed in the Phasighat population (∼0.06%: both leaf & bark). Across the geographic regions, a higher percent of rohitukine was found in the Central-southern Western Ghats, whereas lower in the northern parts of the Western Ghats and Northeast regions. The leaves produce a considerably higher percent of rohitukine and could be used as a sustainable source of rohitukine. The rohitukine analogues, along with other chromone alkaloids of D. binecatariferum were found to be more interactive with the "kinases" family of proteins, majorly "Serine/threonine-protein kinase PFTAIRE-2" (CDK15) with high confidence level (0.94-0.98). The molecular docking of these chromone alkaloids found a strong binding energy with six CDKs (-3.1 to -10.6 kcal/mol) along with a promising ADMET profile. In addition, molecular dynamic simulation found that the rohitukine complexes are virtually constant with CDK-1, 2, 9 and 15, which is substantiated with MM-PBSA free energy calculations. The chromone alkaloids, majorly rohitukine and its analogues were closely clustered with flavopiridol, P-276-00 and IIIM-290 along with other chrotacumines in the chemical phylogeny. In conclusion, D. binectariferum is a rich source of chromone alkaloids, which could lead to the discovery of more potential scaffolding for CDK inhibitors as anticancer drugs.
Collapse
Affiliation(s)
- E Varun
- Center for Ayurveda Biology and Holistic Nutrition, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, 560064, India
| | - K Bhakti
- Center for Ayurveda Biology and Holistic Nutrition, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, 560064, India
| | - K Aishwarya
- Center for Ayurveda Biology and Holistic Nutrition, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, 560064, India
| | - R Hosur Suraj
- College of Forestry, Sirsi, 581401, University of Agricultural Sciences, Dharwad, India
| | - M R Jagadish
- College of Forestry, Sirsi, 581401, University of Agricultural Sciences, Dharwad, India
| | - P Mohana Kumara
- Center for Ayurveda Biology and Holistic Nutrition, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, 560064, India.,Department of Biotechnology and Crop improvement, Kittur Rani Channamma College of Horticulture (KRCCH), Arabhavi, 591218, University of Horticultural Sciences, Bagalkot, India
| |
Collapse
|
37
|
Abstract
Computer-aided drug design (CADD) approaches are playing an increasingly important role in understanding the fundamentals of ligand-receptor interactions and helping medicinal chemists design therapeutics. About 5 years ago, we presented a chapter devoted to an overview of CADD methods and covered typical CADD protocols including structure-based drug design (SBDD) and ligand-based drug design (LBDD) approaches that were frequently used in the antibiotic drug design process. Advances in computational hardware and algorithms and emerging CADD methods are enhancing the accuracy and ability of CADD in drug design and development. In this chapter, an update to our previous chapter is provided with a focus on new CADD approaches from our laboratory and other peers that can be employed to facilitate the development of antibiotic therapeutics.
Collapse
Affiliation(s)
- Wenbo Yu
- Department of Pharmaceutical Sciences, Computer-Aided Drug Design Center, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD, USA.
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD, USA.
| | - David J Weber
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD, USA
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, Computer-Aided Drug Design Center, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD, USA.
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
38
|
Hu Y, Li R, Jin J, Wang Y, Ma R. Quercetin improves pancreatic cancer chemo-sensitivity by regulating oxidative-inflammatory networks. J Food Biochem 2022; 46:e14453. [PMID: 36181395 DOI: 10.1111/jfbc.14453] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 01/14/2023]
Abstract
Chemotherapy is the main method for controlling pancreatic cancer metastasis but the prevalent chemotherapy resistance limits its utilization. The response of oxidation and inflammation often promotes pancreatic cancer progression and chemo-resistance. It is critical to explore the potential natural products with few side effects to control inflammatory responses and understand the related mechanisms. Quercetin is a flavonoid widely found in numerous vegetables, fruits, and foods and is thought to have antioxidant and anti-inflammatory properties, which may be associated with improvement of chemotherapy sensitivity during pancreatic cancer treatment. Quercetin may sensitize pancreatic cancer cells to the chemotherapeutic agents, including bromodomain and extraterminal domain inhibitors (BETI), daunorubicin, gemcitabine, sulforaphane, doxorubicin, and tumor necrosis factor-related signaling apoptosis-inducing ligand (TRAIL). Meanwhile, during the chemo-resistance therapy, many signaling molecules are involved with toll-like receptor 4 (TLR4)-mediated oxidative and inflammatory pathway. The effects of quercetin on other oxidative and inflammatory pathways were also explored. Quercetin may exert antitumor activity during the prevention of pancreatic cancer progression by regulating oxidative and inflammatory networks, which can promote immune escape of cancer cells by inducing immunosuppressive cytokines. Studying these patterns will help us to better understand the functional role of quercetin in the improvement of pancreatic cancer chemo-sensitivity. PRACTICAL APPLICATIONS: Chemotherapy is the major way for treating pancreatic cancer metastasis but the prevalent chemotherapy resistance caused by oxidative and inflammatory responses limits its utilization. It is necessary to explore the potential natural products with few side effects to prevent the oxidative and inflammatory responses. Quercetin is a flavonoid widely found in numerous vegetables, fruits, and foods and is thought to have antioxidant and anti-inflammatory properties, which may be associated with improvement of chemotherapy sensitivity of pancreatic cancer treatment by sensitizing pancreatic cancer cells to various chemotherapeutic agents via the regulation of oxidative and inflammatory networks. Studying these patterns will help us to better understand the functional role of quercetin in the improvement of pancreatic cancer chemo-sensitivity.
Collapse
Affiliation(s)
- Yaoyuan Hu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junyi Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yihui Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Ma
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|