1
|
Chandpa HH, Naskar S, Meena J. Computer Integrated Dominant Epitopes Evoke Protective Immune Response Against Streptococcus pneumoniae. Immunology 2025; 175:180-199. [PMID: 40056072 DOI: 10.1111/imm.13920] [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: 07/11/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 05/07/2025] Open
Abstract
Streptococcus pneumoniae is a gram-positive bacterium responsible for various diseases like pneumonia, acute otitis media, sinusitis, meningitis and bacteraemia. These diseases cause a significant amount of morbidity and mortality. Although polysaccharide vaccines are available, the protection provided by these vaccines is serotype-dependent and not enough in sensitive populations like children and older people. Designing a subunit vaccine by using proteins that are responsible for the pathogenesis of diseases can provide better protection against bacterial infections. In this study, we present the design of a novel multi-epitope vaccine against Streptococcus pneumoniae using an immunoinformatic approach. More than 1170 epitopes were identified against B cells, cytotoxic T lymphocytes and helper T lymphocytes from more than 60 pneumococcal proteins. Epitopes were further screened, and potential epitopes were selected for vaccine development. Seven different vaccine combinations that harbour the 15 dominant B-cell, cytotoxic T cell and helper T cell epitopes were evaluated with linker and β-defensin adjuvant to finalise the best vaccine construct. Bioinformatics tools were used to analyse the construct's physicochemical properties, secondary and tertiary structures, allergenicity, antigenicity and immunogenicity. Docking studies with the TLR-4 receptor and molecular dynamics simulations indicated strong binding affinity and stability. In silico immune response simulations predicted robust IgG immune response generation and observed more than 200 000 IgG1 + IgG2 counts per mL. Similarly, cell-mediated immunity was also enhanced by the designed vaccine construct. The construct was codon-optimised and cloned in silico for expression in Escherichia coli. These findings suggest that the construct is a promising candidate for further experimental validation.
Collapse
Affiliation(s)
- Hitesh Harsukhbhai Chandpa
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Shovan Naskar
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Jairam Meena
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
2
|
Sevim B, Güneş Altuntaş E. Molecular Dynamic Study on the Structure and Thermal Stability of Mutant Pediocin PA-1 Peptides Engineered with Cysteine Substitutions. Probiotics Antimicrob Proteins 2025; 17:1739-1753. [PMID: 38424320 DOI: 10.1007/s12602-024-10225-3] [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] [Accepted: 01/24/2024] [Indexed: 03/02/2024]
Abstract
Pediocin and analogous bacteriocins, valued for thermal stability, serve as versatile antimicrobials in the food sector. Improving their resilience at high temperatures and deriving derivatives not only benefit food production but also offer broad-spectrum antimicrobial potential in pharmaceuticals, spanning treatments for peptic ulcers, women's health, and novel anticancer agents. The study aims to create mutant peptides capable of establishing a third disulfide bond or enhanced through cysteine substitutions. This involves introducing additional Cys residues into the inherent structure of pediocin PA-1 to facilitate disulfide bond formation. Five mutants (Mut 1-5) were systematically generated with double Cys substitutions and assessed for thermal stability through MD simulations across temperatures (298-394 K). The most robust mutants (Mut 1, Mut 4-5) underwent extended analysis via MD simulations, comparing their structural stability, secondary structure, and surface accessibility to the reference Pediocin PA-1 molecule. This comprehensive assessment aims to understand how Cys substitutions influence disulfide bonds and the overall thermal stability of the mutant peptides. In silico analysis indicated that Mut 1 and Mut 5, along with the reference structure, lose their helical structure and one natural disulfide bond at high temperatures, and may impacting antimicrobial activity. Conversely, Mut 4 retained its helical structure and exhibited thermal stability similar to Pediocin PA-1. Pending further experimental validation, this study implies Mut 4 may have high stability and exceptional resistance to high temperatures, potentially serving as an effective antimicrobial alternative.
Collapse
Affiliation(s)
- Büşra Sevim
- Ankara University Biotechnology Institute, Ankara, Turkey
| | | |
Collapse
|
3
|
Schulz L, Ung KL, Zuzic L, Koutnik-Abele S, Schiøtt B, Stokes DL, Pedersen BP, Hammes UZ. Transport of phenoxyacetic acid herbicides by PIN-FORMED auxin transporters. NATURE PLANTS 2025:10.1038/s41477-025-01984-0. [PMID: 40263580 DOI: 10.1038/s41477-025-01984-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/12/2025] [Indexed: 04/24/2025]
Abstract
Auxins are a group of phytohormones that control plant growth and development. Their crucial role in plant physiology has inspired development of potent synthetic auxins that can be used as herbicides. Phenoxyacetic acid derivatives are a widely used group of auxin herbicides in agriculture and research. Despite their prevalence, the identity of the transporters required for distribution of these herbicides in plants is both poorly understood and the subject of controversial debate. Here we show that PIN-FORMED auxin transporters transport a range of phenoxyacetic acid herbicides across the membrane. We go on to characterize the molecular determinants of substrate specificity using a variety of different substrates as well as protein mutagenesis to probe the binding site. Finally, we present cryogenic electron microscopy structures of Arabidopsis thaliana PIN8 bound to either 2,4-dichlorophenoxyacetic acid or 4-chlorophenoxyacetic acid. These structures represent five key states from the transport cycle, allowing us to describe conformational changes associated with the transport cycle. Overall, our results reveal that phenoxyacetic acid herbicides use the same export machinery as endogenous auxins and exemplify how transporter binding sites undergo transformations that dictate substrate specificity. These results provide a foundation for future development of novel synthetic auxins and for precision breeding of herbicide-resistant crop plants.
Collapse
Affiliation(s)
- Lukas Schulz
- Plant Systems Biology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Kien Lam Ung
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lorena Zuzic
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Sarah Koutnik-Abele
- Plant Systems Biology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Birgit Schiøtt
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - David L Stokes
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Bjørn Panyella Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
- Institute for Advanced Study, Technical University of Munich, Garching, Germany.
| | - Ulrich Z Hammes
- Plant Systems Biology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| |
Collapse
|
4
|
Raen R, Islam MM, Islam R, Islam MR, Jarin T. Functional characterization and structural prediction of hypothetical proteins in monkeypox virus and identification of potential inhibitors. Mol Divers 2025; 29:1589-1617. [PMID: 39043911 DOI: 10.1007/s11030-024-10935-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
The excessive activation of the monkeypox virus (MPXV-Congo_8-156) is linked to various skin and respiratory disorders such as rashes, fluid-filled blisters, swollen lymph nodes and encephalitis (inflammation of the brain), highlighting MPXV-Congo_8-156 as a promising target for drug intervention. Despite the effectiveness of Cidofovir, in inhibiting MPXV activity, its limited ability to penetrate the skin and its strong side effects restrict its application. To address this challenge, we screened 500 compounds capable of penetrating the skin and gastrointestinal tract to identify potent MPXV inhibitors. Various characterization schemes and structural models of MPXV-Congo_8-156 were explored with bioinformatics tools like PROTPARAM, SOPMA, SWISS-MODEL and PROCHECK. Using molecular docking in PyRx, we evaluated the binding affinities of these compounds with MPXV-Congo_8-156 and identified the top five candidates ranging from - 9.2 to - 8.8 kcal/mol. ADMET analysis indicated that all five compounds were safer alternatives, showing no AMES toxicity or carcinogenicity in toxicological assessments. Molecular dynamics (MD) simulations, conducted for 100 ns each, confirmed the docking interactions of the top five compounds alongside the control (Cidofovir), validating their potential as MPXV inhibitors. The compounds with PubChem CID numbers 4061636, 4422538, 3583576, 4856107 and 4800629 demonstrated strong support in terms of root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), radius of gyration (Rg), solvent-accessible surface area (SASA) value, hydrogen bond analysis, and Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) analysis. Thus, our investigation identified these five compounds as promising inhibitors of MPXV, offering potential therapeutic avenues. However, further in vivo studies are necessary to validate our findings.
Collapse
Affiliation(s)
- Reana Raen
- Department of Biomedical Engineering, Khulna University of Engineering & Technology, Khulna, Bangladesh.
- Department of Biomedical Engineering, Chittagong University of Engineering & Technology, Chittagong, Bangladesh.
| | - Muhammad Muinul Islam
- Department of Biomedical Engineering, Khulna University of Engineering & Technology, Khulna, Bangladesh
| | - Redwanul Islam
- Department of Biomedical Engineering, Khulna University of Engineering & Technology, Khulna, Bangladesh
| | - Md Rabiul Islam
- Department of Electrical and Electronic Engineering, Jashore University of Science & Technology, Jashore, Bangladesh
| | - Tanima Jarin
- Department of Agriculture, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| |
Collapse
|
5
|
Motiwala T, Nyide B, Khoza T. Molecular dynamic simulations to assess the structural variability of ClpV from Enterobacter cloacae. FRONTIERS IN BIOINFORMATICS 2025; 5:1498916. [PMID: 40201065 PMCID: PMC11975955 DOI: 10.3389/fbinf.2025.1498916] [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: 09/19/2024] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
The Enterobacter cloacae complex (ECC) consists of six Enterobacter species (E. cloacae, hormaechei, kobei, ludwigii, nimipressuralis and asburiae) that have emerged as nosocomial pathogens of interest, with E. cloacae and Enterobacter hormachei being the most frequently isolated ECC species in human clinical specimens and intensive care unit (ICU) patients. Many nosocomial outbreaks of E. cloacae have been related to transmission through contaminated surgical equipment and operative cleaning solutions. As this pathogen evades the action of antibiotics, it is important to find alternative targets to limit the devastating effects of these pathogens. ClpV is a Clp ATPase which dissociates and recycles the contracted sheath of the bacterial type VI secretion system (T6SS), thereby regulating bacterial populations and facilitating environmental colonization. Seventy-one Enterobacter strains were mined for Clp ATPase proteins. All the investigated strains contained ClpA, ClpB, ClpX and ClpV while only 20% contained ClpK. All the investigated strains contained more than one ClpV protein, and the ClpV proteins showed significant variations. Three ClpV proteins from E. cloacae strain E3442 were then investigated to determine the structural difference between each protein. Homology modelling showed the proteins to be structurally similar to each other, however the physicochemical characteristics of the proteins vary. Additionally, physicochemical analysis and molecular dynamic simulations showed that the proteins were highly dynamic and not significantly different from each other. Further investigation of the proteins in silico and in vitro in the presence and absence of various ligands and proteins could be performed to determine whether the proteins all interact with their surroundings in the same manner. This would allow one to determine why multiple homologs of the same protein are expressed by pathogens.
Collapse
Affiliation(s)
| | | | - Thandeka Khoza
- Department of Biochemistry, School of Life Sciences, Pietermaritzburg Campus, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| |
Collapse
|
6
|
Prottay AAS, Emamuzzaman, Ripu TR, Sarwar MN, Rahman T, Ahmmed MS, Bappi MH, Emon M, Ansari SA, Coutinho HDM, Islam MT. Anxiogenic-like effects of coumarin, possibly through the GABAkine interaction pathway: Animal studies with in silico approaches. Behav Brain Res 2025; 480:115392. [PMID: 39667645 DOI: 10.1016/j.bbr.2024.115392] [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: 07/06/2024] [Revised: 09/18/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Anxiety disorder is the most common mental illness and a major contributor to impairment. Thus, there is an urgent need to find novel lead compounds to mitigate anxiety. It is widely recognized that the neurobiology of anxiety-related behavior involves GABAergic systems. OBJECTIVES This research aimed to examine the anxiogenic action of coumarin (CMN), a natural benzopyrone derived from plants, and determine its underlying mechanism through in vivo and in silico investigations. METHODS This was accomplished by using a variety of behavioral procedures, including open field, swing, hole cross, and light-dark tests, on male and female Swiss albino mice that had been orally administered three experimental doses of CMN (1, 2, and 4 mg/kg). The CMN group was also examined with the GABAA receptor agonist diazepam (DZP, 2 mg/kg) and flumazenil antagonist (FLU, 0.1 mg/kg). Furthermore, CMN and standards were subjected to a molecular docking analysis to determine their binding affinities for the GABAA receptor subunits (α1, α4, β2, γ2, and δ). Several software programs were used to visualize the ligand-receptor interaction and analyze the pharmacokinetic profile. RESULTS Compared to typical treatments, our results show that CMN (1 mg/kg) significantly (p < 0.05) increases the locomotor activity of animals. Furthermore, CMN exerted the highest binding affinity (-6.5 kcal/mol) with the GABA-α1 receptor compared to conventional DZP. Along with FLU, CMN displayed several hydrophobic and hydrogen bonds with GABAA receptor subunits. The pharmacokinetic and drug-like properties of CMN are also remarkable. In animal studies, CMN worked synergistically with FLU to provide anxiogenic-like effects. CONCLUSION We conclude that, based on in vivo and in silico data, CMN, alone or in combination with FLU, may be employed in future neurological clinical studies. However, further research is needed to confirm this behavioral activity and elucidate the possible mechanism of action.
Collapse
Affiliation(s)
- Abdullah Al Shamsh Prottay
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
| | - Emamuzzaman
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
| | - Tawfik Rakaiyat Ripu
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md Nazim Sarwar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Towfiqur Rahman
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md Shakil Ahmmed
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Mehedi Hasan Bappi
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Md Emon
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, Brazil.
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh; Pharmacy Discipline, Khulna University, Khulna 9208, Bangladesh.
| |
Collapse
|
7
|
Yadav JK, Ghanchi M, Dixit N, Sindhav G, Patel S, Rawal R. Phytonutrients as a Defensive Barrier Against G Ectodomain Fusion in Chandipura Virus Infection. Mol Biotechnol 2025:10.1007/s12033-025-01384-x. [PMID: 39998775 DOI: 10.1007/s12033-025-01384-x] [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: 10/10/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025]
Abstract
Viruses, microscopic menace that transcends time leaving its mark on every era have been silent predators since the dawn of civilization, evolving with us and shaping our history. Chandipura virus (CHPV), a potent member of the Rhabdoviridae family poses a significant threat in India with rapid neuroinvasive potential leading to fatal encephalitis, particularly in children. Given the scarcity of research, our study consolidates critical information regarding its lifecycle, fusion process, and reviewed the LRP1 and GRP78 as CHPV target receptors. With no FDA-approved drugs currently available for CHPV prevention, our research focuses on identifying potential molecules that can disrupt the virus at its most critical juncture, the fusion stage. The results derived from compounds screening indicated Silibinin, 3-(2,3-Dihydroxy-3-Methylbutyl)-6-Hydroxy-2-[(1E,5E)-3,4,10-Trihydroxyundeca-1,5-Dienyl] Benzaldehyde, Budmunchiamine L5, and L4 as a leading molecule may efficaciously inhibit G ectodomain fusion. By analyzing pharmacokinetic properties through radar graph, outcomes support the nomination of four compounds as potential inhibitory molecules and ensure they possess the optimal balance of drug-like characteristics. Working with the CHPV presents significant challenges, making the in silico parameters crucial in guiding future research. Our study sought to pioneer the discovery of therapeutic molecules against the CHPV, providing a foundational framework for developing effective antiviral strategies.
Collapse
Affiliation(s)
- Jyoti Kumari Yadav
- Department of Life Sciences, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Mohammadfesal Ghanchi
- Department of Zoology, BMT, HGC and WBC, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Nandan Dixit
- Department of Botany, Bioinformatics and Climate Change Impacts Management, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Gaurang Sindhav
- Department of Zoology, BMT, HGC and WBC, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Saumya Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Rakesh Rawal
- Department of Life Sciences, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, 380009, India.
- Department of Medical Biotechnology, Gujarat Biotechnology University, GIFT City, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
8
|
Disha IJ, Hasan R, Bhuia S, Ansari SA, Ansari IA, Islam MT. Anxiolytic Efficacy of Indirubin: In Vivo Approach Along with Receptor Binding Profiling and Molecular Interaction with GABAergic Pathways. ChemistryOpen 2025; 14:e202400290. [PMID: 39460441 PMCID: PMC11808267 DOI: 10.1002/open.202400290] [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: 08/06/2024] [Revised: 09/03/2024] [Indexed: 10/28/2024] Open
Abstract
Anxiety is a natural response to stress, characterized by feelings of worry, fear, or unease. The current research was conducted to investigate the anxiolytic effect of indirubin (IND) in different behavioral paradigms in Swiss albino mice. To observe the animal's behavioural response to assess anxiolytic activity, different tests were performed, such as the open-field (square cross, grooming, and rearing), swing, dark-light, and hole cross tests. The experimental mice were administered IND (5 and 10 mg/kg, p.o.), where diazepam (DZP) and vehicle were used as positive and negative controls, respectively. In addition, a combination treatment (DZP+IND-10) was provided to the animals to determine the modulatory effect of IND on DZP. Molecular docking approach was also conducted to determine the binding energy of IND with the GABAA receptor (α2 and α3 subunits) and pharmacokinetics were also estimated. The findings revealed that IND dose-dependently significantly (p<0.05) reduced the animal's movement exerting calming behavior like DZP. IND also demonstrated the highest docking score (-7.7 kcal/mol) against the α3 subunit, while DZP showed a lower docking value (-6.4 kcal/mol) than IND. The ADMET analysis revealed that IND has proper drug-likeness and pharmacokinetic characteristics. In conclusion, IND exerted anxiolytic effects through GABAergic Pathways.
Collapse
Affiliation(s)
- Ishrat Jahan Disha
- Biochemistry and Molecular BiologyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganj8100Bangladesh
- Bioinformatics and Drug Innovation LaboratoryBioLuster Research Center Ltd.Gopalganj, Dhaka8100Bangladesh
| | - Rubel Hasan
- Bioinformatics and Drug Innovation LaboratoryBioLuster Research Center Ltd.Gopalganj, Dhaka8100Bangladesh
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganj8100Bangladesh
| | - Shimul Bhuia
- Bioinformatics and Drug Innovation LaboratoryBioLuster Research Center Ltd.Gopalganj, Dhaka8100Bangladesh
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganj8100Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical ChemistryCollege of PharmacyKing Saud UniversityRiyadh11451Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and TechnologyUniversity of TurinTurin10124Italy
| | - Muhammad Torequl Islam
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganj8100Bangladesh
- Pharmacy DisciplineKhulna UniversityKhulna9208Bangladesh
| |
Collapse
|
9
|
Zafar S, Bai Y, Muhammad SA, Guo J, Khurram H, Zafar S, Muqaddas I, Shaikh RS, Bai B. Molecular dynamics simulation based prediction of T-cell epitopes for the production of effector molecules for liver cancer immunotherapy. PLoS One 2025; 20:e0309049. [PMID: 39752339 PMCID: PMC11698456 DOI: 10.1371/journal.pone.0309049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/05/2024] [Indexed: 01/06/2025] Open
Abstract
Liver cancer is the sixth most frequent malignancy and the fourth major cause of deaths worldwide. The current treatments are only effective in early stages of cancer. To overcome the therapeutic challenges and exploration of immunotherapeutic options, broad spectral therapeutic vaccines could have significant impact. Based on immunoinformatic and integrated machine learning tools, we predicted the potential therapeutic vaccine candidates of liver cancer. In this study, machine learning and MD simulation-based approach are effectively used to design T-cell epitopes that aid the immune system against liver cancer. Antigenicity, molecular weight, subcellular localization and expression site predictions were used to shortlist liver cancer associated proteins including AMBP, CFB, CDHR5, VTN, APOBR, AFP, SERPINA1 and APOE. We predicted CD8+ T-cell epitopes of these proteins containing LGEGATEAE, LLYIGKDRK, EDIGTEADV, QVDAAMAGR, HLEARKKSK, HLCIRHEMT, LKLSKAVHK, EQGRVRAAT and CD4+ T-cell epitopes of VLGEGATEA, WVTKQLNEI, VEEDTKVNS, FTRINCQGK, WGILGREEA, LQDGEKIMS, VKFNKPFVF, VRAATVGSL. We observed the substantial physicochemical properties of these epitopes with a significant binding affinity with MHC molecules. A polyvalent construct of these epitopes was designed using suitable linkers and adjuvant indicated significant binding energy (>-10.5 kcal/mol) with MHC class-I and II molecule. Based on in silico cloning, we found the considerable compatibility of this polyvalent construct with the E. coli expression system and the efficiency of its translation in host. The system-level and machine learning based cross validations showed the possible effect of these T-cell epitopes as potential vaccine candidates for the treatment of liver cancer.
Collapse
Affiliation(s)
- Sidra Zafar
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Multan, Punjab, Pakistan
| | - Yuhe Bai
- Department of Computer Science, Sorbonne University, Paris, France
| | - Syed Aun Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Multan, Punjab, Pakistan
| | - Jinlei Guo
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, Henan, China
| | - Haris Khurram
- Department of Mathematics and Computer Science, Faculty of Science and Technology, Prince of Songkla University, Pattani Campus, Pattani, Thailand
- Department of Sciences and Humanities, National University of Computer and Emerging Sciences, Chiniot, Punjab, Pakistan
| | - Saba Zafar
- Department of Biochemistry and Biotechnology, The Women University Multan, Multan, Punjab, Pakistan
| | - Iraj Muqaddas
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Multan, Punjab, Pakistan
| | - Rehan Sadiq Shaikh
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Punjab, Pakistan
| | - Baogang Bai
- School of Information and Technology, Wenzhou Business College, Wenzhou, Zhejiang, China
- Zhejiang Province Engineering Research Center of Intelligent Medicine, Wenzhou, China
- The 1 School of Medical, School of Information and Engineering, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Jaiswal A, Pandey AK, Tripathi A, Dubey SK. Omics-centric evidences of fipronil biodegradation by Rhodococcus sp. FIP_B3. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 364:125320. [PMID: 39549993 DOI: 10.1016/j.envpol.2024.125320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
The widespread use of the pesticide fipronil in domestic and agriculture sectors has resulted in its accumulation across the environment. Its use to assure food security has inadvertently affected soil microbiome composition, fertility and, ultimately, human health. Degradation of residual fipronil present in the environment using specific microbial species is a promising strategy for its removal. The present study delves into the omics approach for fipronil biodegradation using the native bacterium Rhodococcus sp. FIP_B3. It has been observed that within 40 days, nearly 84% of the insecticide gets degraded. The biodegradation follows a pseudo-first-order kinetics (k = 0.0197/d with a half-life of ∼11 days). Whole genome analysis revealed Cytochrome P450 monooxygenase, peroxidase-related enzyme, haloalkane dehalogenase, 2-nitropropane dioxygenase, and aconitate hydratase are involved in the degradation process. Fipronil-sulfone, 5-amino-1-(2-chloro-4-(trifluoromethyl)phenyl)-4- ((trifluoromethyl)sulfonyl)-1H-pyrazole-3-carbonitrile, (E)-5-chloro-2-oxo-3- (trifluoromethyl)pent-4-enoic acid, 4,4,4-trifluoro-2-oxobutanoic acid, and 3,3,3- trifluoropropanoic acid were identified as the major metabolites that support the bacterial degradation of fipronil. In-silico molecular docking and molecular dynamic simulation-based analyses of degradation pathway intermediates with their respective enzymes have indicated stable interactions with significant binding energies (-5.9 to -9.7 kcal/mol). These results have provided the mechanistic cause of the elevated potential of Rhodococcus sp. FIP_B3 for fipronil degradation and will be advantageous in framing appropriate strategies for the bioremediation of fipronil-contaminated environment.
Collapse
Affiliation(s)
- Anjali Jaiswal
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi- 221005, India
| | - Anand Kumar Pandey
- Department of Biotechnology Engineering, Institute of Engineering and Technology, Bundelkhand University, Jhansi, 284128, India
| | - Animesh Tripathi
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi- 221005, India
| | - Suresh Kumar Dubey
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi- 221005, India.
| |
Collapse
|
11
|
Yang W, Ye C, Wang L, Nie J, Liu X, Zhang T, Zhang W, Saba NU, Yin L, Xing L, Su X. Binding properties of olfactory proteins to host volatiles, free fatty acids and cuticular hydrocarbons in the termite Reticulitermes aculabialis. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 176:104211. [PMID: 39542378 DOI: 10.1016/j.ibmb.2024.104211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/25/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
As eusocial insects prevalent in tropical and subtropical regions, termites are characterized by highly organized behaviors and exceptional adaptability, rooted in caste differentiation and chemical communication. These traits make them excellent models for studying insect social structures and ecological interactions. Investigating how termites use chemical signals to perceive and respond to their environment provides insights into their coordination and adaptation within complex ecosystems. This study delved into the chemosensory mechanisms of Reticulitermes aculabialis, examining the interactions of four olfactory proteins with 70 ligands, including host volatiles, cuticular hydrocarbons (CHCs), and free fatty acids (FFAs). Molecular docking simulations revealed varied affinities of the olfactory proteins for long-chain hydrocarbons (n-C23 to n-C28), suggesting a nuanced chemical communication system through specific hydrocarbon detection. RacuCSP1 and RacuCSP2 exhibited specific binding to linoleic acid and undecanoic acid, respectively, highlighting the significance of FFAs in the physiological and behavioral processes of termites. The four olfactory proteins showed a strong affinity for longifolene in fluorescence competitive binding experiments. Notably, RacuOBPs exhibited unique affinities for terpenoid volatiles such as β-lonone and neocembrene, while RacuCSPs specifically bound with terpenoids like 3-carene, myrtenol, α-pinene oxide and β-pinene indicating their critical roles in host detection. Behavioral observations following gene silencing revealed that RacuOBP5 was essential for recognizing longifolene and α-lonone recognition, while RacuCSP1 was key for detecting α-pinene in termites. These findings enhance our understanding of the termite chemosensory system and offer insights for developing precise pest management strategies.
Collapse
Affiliation(s)
- Wenxu Yang
- College of Life Sciences, Northwest University, Xi'an, China
| | - Chenxu Ye
- College of Life Sciences, Northwest University, Xi'an, China
| | - Lu Wang
- College of Life Sciences, Northwest University, Xi'an, China
| | - Jinjuan Nie
- College of Life Sciences, Northwest University, Xi'an, China
| | - Xinyi Liu
- College of Life Sciences, Northwest University, Xi'an, China
| | - Tiange Zhang
- College of Life Sciences, Northwest University, Xi'an, China
| | - Wenxiu Zhang
- College of Life Sciences, Northwest University, Xi'an, China
| | - Noor Us Saba
- College of Life Sciences, Northwest University, Xi'an, China
| | - Lingfang Yin
- College of Life Sciences, Northwest University, Xi'an, China
| | - Lianxi Xing
- College of Life Sciences, Northwest University, Xi'an, China
| | - Xiaohong Su
- College of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
12
|
Sah SN, Gupta S, Bhardwaj N, Gautam LK, Capalash N, Sharma P. In silico design and assessment of a multi-epitope peptide vaccine against multidrug-resistant Acinetobacter baumannii. In Silico Pharmacol 2024; 13:7. [PMID: 39726905 PMCID: PMC11668725 DOI: 10.1007/s40203-024-00292-3] [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: 08/21/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Acinetobacter baumannii, an opportunistic and notorious nosocomial pathogen, is responsible for many infections affecting soft tissues, skin, lungs, bloodstream, and urinary tract, accounting for more than 722,000 cases annually. Despite the numerous advancements in therapeutic options, no approved vaccine is currently available for this particular bacterium. Consequently, this study focused on creating a rational vaccine design using bioinformatics tools. Three outer membrane proteins with immunogenic potential and properties of good vaccine candidates were used to select epitopes based on good antigenic properties, non-allergenicity, high binding scores, and a low IC50 value. A multi-epitope peptide (MEP) construct was created by sequentially linking the epitopes using suitable linkers. ClusPro 2.0 and C-ImmSim web servers were used for docking analysis with TLR2/TLR4 and immune response respectively. The Ramachandran plot showed an accurate model of the MEP with 100% residue in the most favored and allowed regions. The construct was highly antigenic, stable, non-allergenic, non-toxic, and soluble, and showed maximum population coverage. Additionally, molecular docking demonstrated strong binding between the designed MEP vaccine and TLR2/TLR4. In silico immunological simulations showed significant increases in T-cell and B-cell populations. Finally, codon optimization and in silico cloning were conducted using the pET-28a (+) plasmid vector to evaluate the efficiency of the expression of vaccine peptide in the host organism (Escherichia coli). This designed MEP vaccine would support and accelerate the laboratory work to develop a potent vaccine targeting MDR Acinetobacter baumannii. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00292-3.
Collapse
Affiliation(s)
- Shiv Nandan Sah
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
- Department of Microbiology, Central Campus of Technology, Tribhuvan University, Dharan, Nepal
| | - Sumit Gupta
- School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, 110062 India
| | - Neha Bhardwaj
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
| | - Lalit Kumar Gautam
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242 USA
- Department of Biotechnology, Panjab University, Chandigarh, 160014 India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, 160014 India
| | - Prince Sharma
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
| |
Collapse
|
13
|
Seifeldin S, Saeed M, Alshaghdali K, Yousif E, Abu Sabaa A, Rabie H, Siddiqui S, Saeed A. Investigating the effects of the ARG258HIS mutation on RAD51C in inherited Fanconi Anemia and cancer disease. J Biomol Struct Dyn 2024:1-11. [PMID: 39648652 DOI: 10.1080/07391102.2024.2431656] [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: 01/21/2024] [Accepted: 05/03/2024] [Indexed: 12/10/2024]
Abstract
Fanconi anemia is a rare chromosomal instability disorder associated with developmental abnormalities, bone marrow failure, and a heightened susceptibility to leukemia and other cancers. It is an autosomal recessive genetic disorder, necessitating both parents to carry the faulty gene. Diagnostic methods include blood tests, chromosome breakage assessments, and genetic testing. While there is no cure, treatments encompass blood transfusions, bone marrow transplants, and gene therapy, with patients requiring regular check-ups, supportive care, and cancer screening to enhance their quality of life. In this study, we identify a specific substitution (R258H) targeting the crucial binding site of the alpha-helix region in RAD51C. This substitution induces structural disorder in distinct regions, as indicated by the near absence of electron density for multiple amino acids. Intriguingly, these disordered regions do not follow a continuous sequence from the mutation site and extend across domain boundaries. We utilized computational prediction algorithms and Molecular Dynamics (MD) simulations to model RAD51C and its mutation (R258H) structurally. These simulations highlighted alterations in conformational dynamics, the Free Energy Landscape (FEL), and intrinsic molecular motions induced by the mutation, suggesting structural destabilization that could disrupt its function. This observed destabilization in RAD51C due to mutations offers valuable insights that may serve as diagnostic markers for individuals carrying these mutations, particularly in Fanconi anemia.
Collapse
Affiliation(s)
- Sara Seifeldin
- Department of Clinical Laboratory Science, College of Applied Medical Science, University of Hail, Hail, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Khalid Alshaghdali
- Department of Clinical Laboratory Science, College of Applied Medical Science, University of Hail, Hail, Saudi Arabia
| | - Elgeli Yousif
- Department of Diagnostic Radiology, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Amal Abu Sabaa
- Department of Immunology, Genetics & Pathology, Uppsala University, Sweden
| | - Hatem Rabie
- Ministry of Health -Hail Regional Laboratory, Hail, Saudi Arabia
| | - Samra Siddiqui
- Department Health Services Management, College of Public Health and Health Informatics, University of Hail, Hail, Saudi Arabia
| | - Amir Saeed
- Department of Clinical Laboratory Science, College of Applied Medical Science, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
14
|
Andrade Júnior FPD, Galdino Gouveia R, Ilan Soares Medeiros C, Teixeira BDA, Farias BKDS, Oliveira NDR, Silva DDF, Lima EDO. Antifungal activity of citronellal against Trichophyton rubrum and its predictive mechanism of action by CYP51 inhibition through molecular docking. Nat Prod Res 2024; 38:4125-4133. [PMID: 37933528 DOI: 10.1080/14786419.2023.2277352] [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: 08/18/2023] [Revised: 10/03/2023] [Accepted: 10/22/2023] [Indexed: 11/08/2023]
Abstract
The present study aimed to investigate the antifungal activity of citronellal (CIT) against clinical isolates of T. rubrum and to show the possible mechanism of action involved. The antifungal potential of CIT was evaluated from the Minimum Inhibitory Concentration (MIC), Minimum Fungicide Concentration (MFC) and assays with ergosterol and sorbitol, to elucidate the possible mechanisms of action, and molecular docking. MIC and MFC values ranged from 4 to 512 µg/mL. Regarding the mechanism of action, the monoterpene demonstrated interaction with fungal ergosterol. In addition, it is possible to observe that CIT acts on crucial enzymes for the biosynthesis and maintenance of the fungal cell membrane, due to the ability of the monoterpene to bind to CYP51. The results obtained in this research demonstrate that CIT has the potential to become, in the future, a product for the treatment of dermatophytosis.
Collapse
Affiliation(s)
| | | | | | - Bráulio de Almeida Teixeira
- Master in Natural and Synthetic Bioactive Products, Federal University of Paraiba (UFPB), João Pessoa, Brazil
| | | | - Nayana da Rocha Oliveira
- Master in Natural and Synthetic Bioactive Products, Federal University of Paraiba (UFPB), João Pessoa, Brazil
| | | | | |
Collapse
|
15
|
Jain H, Rawal E, Kumar P, Sain SK, Siwach P. In Silico Investigation of the Interactions Between Cotton Leaf Curl Multan Virus Proteins and the Transcriptional Gene Silencing Factors of Gossypium hirsutum L. J Mol Evol 2024; 92:891-911. [PMID: 39542922 DOI: 10.1007/s00239-024-10216-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
The highly dynamic nature of the Cotton leaf curl virus (CLCuV) complex (causing Cotton leaf curl disease, a significant global threat to cotton) presents a formidable challenge in unraveling precise molecular mechanisms governing viral-host interactions. To address this challenge, the present study investigated the molecular interactions of 6 viral proteins (Rep, TrAP, C4, C5, V2, and βC1) with 18 cotton Transcriptional Gene Silencing (TGS) proteins. Protein-protein dockings conducted for different viral-host protein pairs using Clustered Protein Docking (ClusPro) and Global RAnge Molecular Matching (GRAMM) (216 docking runs), revealed variable binding energies. The interacting pairs with the highest binding affinities were further scrutinized using bioCOmplexes COntact MAPS (COCOMAPS) server, which revealed robust binding of three viral proteins- TrAP, C4, and C5 with 14 TGS proteins, identifying several novel interactions (not reported yet by earlier studies), such as TrAP targeting DCL3, HDA6, and SUVH6; C4 targeting RAV2, CMT2, and DMT1; and C5 targeting CLSY1, RDR1, RDR2, AGO4, SAMS, and SAHH. Visualizing these interactions in PyMol provided a detailed insight into interacting regions. Further assessment of the impact of 18 variants of the C4 protein on interaction with CMT2 revealed no correlation between sequence variation and docking energies. However, conserved residues in the C4 binding regions emerged as potential targets for disrupting viral integrity. Hence, this study provides valuable insights into the viral-host interplay, advancing our understanding of Cotton leaf curl Multan virus pathogenicity and opening novel avenues for devising various antiviral strategies by targeting the host-viral interacting regions after experimental validation.
Collapse
Affiliation(s)
- Heena Jain
- Department of Biotechnology, Chaudhary Devi Lal University, Sirsa, Haryana, 125055, India
| | - Ekta Rawal
- Department of Biotechnology, Chaudhary Devi Lal University, Sirsa, Haryana, 125055, India
| | - Prabhat Kumar
- Department of Biotechnology, Chaudhary Devi Lal University, Sirsa, Haryana, 125055, India
| | - Satish Kumar Sain
- ICAR-Central Institute of Cotton Research, Regional Station-Sirsa, Sirsa, Haryana, 125055, India
| | - Priyanka Siwach
- Department of Biotechnology, Chaudhary Devi Lal University, Sirsa, Haryana, 125055, India.
| |
Collapse
|
16
|
Mahalingan KK, Grotjahn DA, Li Y, Lander GC, Zehr EA, Roll-Mecak A. Structural basis for α-tubulin-specific and modification state-dependent glutamylation. Nat Chem Biol 2024; 20:1493-1504. [PMID: 38658656 PMCID: PMC11529724 DOI: 10.1038/s41589-024-01599-0] [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: 08/01/2023] [Accepted: 03/06/2024] [Indexed: 04/26/2024]
Abstract
Microtubules have spatiotemporally complex posttranslational modification patterns. Tubulin tyrosine ligase-like (TTLL) enzymes introduce the most prevalent modifications on α-tubulin and β-tubulin. How TTLLs specialize for specific substrate recognition and ultimately modification-pattern generation is largely unknown. TTLL6, a glutamylase implicated in ciliopathies, preferentially modifies tubulin α-tails in microtubules. Cryo-electron microscopy, kinetic analysis and single-molecule biochemistry reveal an unprecedented quadrivalent recognition that ensures simultaneous readout of microtubule geometry and posttranslational modification status. By binding to a β-tubulin subunit, TTLL6 modifies the α-tail of the longitudinally adjacent tubulin dimer. Spanning two tubulin dimers along and across protofilaments (PFs) ensures fidelity of recognition of both the α-tail and the microtubule. Moreover, TTLL6 reads out and is stimulated by glutamylation of the β-tail of the laterally adjacent tubulin dimer, mediating crosstalk between α-tail and β-tail. This positive feedback loop can generate localized microtubule glutamylation patterns. Our work uncovers general principles that generate tubulin chemical and topographic complexity.
Collapse
Affiliation(s)
- Kishore K Mahalingan
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Danielle A Grotjahn
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute La Jolla, La Jolla, CA, USA
| | - Yan Li
- Proteomics Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Gabriel C Lander
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute La Jolla, La Jolla, CA, USA
| | - Elena A Zehr
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
- Biochemistry & Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
17
|
Eskandari A, Leow TC, Rahman MBA, Oslan SN. Structural investigation, computational analysis, and theoretical cryoprotectant approach of antifreeze protein type IV mutants. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:385-403. [PMID: 39327310 DOI: 10.1007/s00249-024-01719-7] [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: 12/16/2023] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 09/28/2024]
Abstract
Antifreeze proteins (AFPs) have unique features to sustain life in sub-zero environments due to ice recrystallization inhibition (IRI) and thermal hysteresis (TH). AFPs are in demand as agents in cryopreservation, but some antifreeze proteins have low levels of activity. This research aims to improve the cryopreservation activity of an AFPIV. In this in silico study, the helical peptide afp1m from an Antarctic yeast AFP was modeled into a sculpin AFPIV, to replace each of its four α-helices in turn, using various computational tools. Additionally, a new linker between the first two helices of AFPIV was designed, based on a flounder AFPI, to boost the ice interaction activity of the mutants. Bioinformatics tools such as ExPASy Prot-Param, Pep-Wheel, SOPMA, GOR IV, Swiss-Model, Phyre2, MODFOLD, MolPropity, and ProQ were used to validate and analyze the structural and functional properties of the model proteins. Furthermore, to evaluate the AFP/ice interaction, molecular dynamics (MD) simulations were executed for 20, 100, and 500 ns at various temperatures using GROMACS software. The primary, secondary, and 3D modeling analysis showed the best model for a redesigned antifreeze protein (AFP1mb, with afp1m in place of the fourth AFPIV helix) with a QMEAN (Swiss-Model) Z score value of 0.36, a confidence of 99.5%, a coverage score of 22%, and a p value of 0.01. The results of the MD simulations illustrated that AFP1mb had more rigidity and better ice interactions as a potential cryoprotectant than the other models; it also displayed enhanced activity in limiting ice growth at different temperatures.
Collapse
Affiliation(s)
- Azadeh Eskandari
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Enzyme Technology and X-Ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | | | - Siti Nurbaya Oslan
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Enzyme Technology and X-Ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
18
|
Shovon MHJ, Imtiaz M, Biswas P, Tareq MMI, Zilani MNH, Hasan M. A pan-genomic analysis based multi-epitope vaccine development by targeting Stenotrophomonas maltophilia using reverse vaccinology method: an in-silico approach. In Silico Pharmacol 2024; 12:93. [PMID: 39464855 PMCID: PMC11499521 DOI: 10.1007/s40203-024-00271-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
Antibiotic resistance in bacteria leads to high mortality rates and healthcare costs, a significant concern for public health. A colonizer of the human respiratory system, Stenotrophomonas maltophilia is frequently associated with hospital-acquired infections in individuals with cystic fibrosis, cancer, and other chronic illnesses. The importance of this study is underscored by its capacity to meet the critical demand for effective preventive strategies against this pathogen, particularly among susceptible groups of cystic fibrosis and those undergoing cancer treatment. In this study, we engineered a multi-epitope vaccine targeting S. maltophilia through genomic analysis, reverse vaccination strategies, and immunoinformatic techniques by examining a total of 81 complete genomes of S. maltophilia strains. Our investigation revealed 1945 core protein-coding genes alongside their corresponding proteomic sequences, with 191 of these genes predicted to exhibit virulence characteristics. Out of the filtered proteins, three best antigenic proteins were selected for epitope prediction while seven epitopes each from CTL, HTL, and B cell were chosen for vaccine development. The vaccine was refined and validated, showing highly antigenic and desirable physicochemical features. Molecular docking assessments revealed stable binding with TLR-4. Molecular dynamic simulation demonstrated stable dynamics with minor alterations. The originality of this investigation is rooted in the thorough techniques aimed at designing a vaccine that directly targets S. maltophilia, a microorganism of considerable clinical relevance that currently lacks an available vaccine. This study not only responds to a pressing public health crisis but also lays the groundwork for subsequent research endeavors focused on the prevention of S. maltophilia outbreaks. Further evidence from studies in mice models is needed to confirm immune protection against S. maltophilia.
Collapse
Affiliation(s)
- Md. Hasan Jafre Shovon
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Md. Imtiaz
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Md. Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Md Nazmul Hasan Zilani
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Md.Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| |
Collapse
|
19
|
Pires ECF, da Silva FP, Schallenberger K, Hermann BS, Mallmann L, Moura WS, Ascêncio SD, Barbosa RDS, Soares IM, Fleck JD, de Oliveira EE, Smagghe G, Ribeiro BM, Aguiar RWDS. Antiviral Potential of Chiococca alba (L.) Hitchc. Plant Extracts Against Chikungunya and Mayaro Viruses. Int J Mol Sci 2024; 25:11397. [PMID: 39518948 PMCID: PMC11546558 DOI: 10.3390/ijms252111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Chikungunya and Mayaro fevers are viral infectious diseases characterized by fever and arthralgia, for which there are currently no effective vaccines or treatments. The urgent need for novel antiviral agents against Chikungunya virus (CHIKV) and Mayaro virus (MAYV) has led to interest in plant-based compounds that can disrupt the viral replication cycle. Chiococca alba (L.) Hitchc., a Neotropical plant traditionally used by Yucatec Maya healers as an antipyretic and antirheumatic, may hold potential as a source of antiviral agents. This study aimed to evaluate the antiviral potential of C. alba methanolic extracts (CAH21 and CAH24) against CHIKV and MAYV through preliminary in vitro and in silico analyses. The cytotoxicity of two methanolic extracts from C. alba roots was assessed in Vero cells using the neutral red assay, and their viral activity was determined via plaque assay post-treatment. Given the observed antiviral effects, we used computational predictions to explore interactions between the multifunctional nsP2 proteases and secondary metabolites identified in C. alba extracts. The metabolites were identified using high-performance liquid chromatography (HPLC) and gas chromatography-mass spectrometry (GC-MS). Phytochemical analysis revealed the presence of flavonoids, coumarins, and phenolic acids in the C. alba extracts. In vitro assays demonstrated that both extracts inhibited over 70% of activity against CHIKV and MAYV at a concentration of 60 µg/mL. In silico predictions suggested that the flavonoids naringin and vitexin had the highest affinity for the nsP2 proteases of CHIKV and MAYV, indicating their potential as viral inhibitors. Our findings revealed that C. alba extract represents a promising source of novel antiviral compounds.
Collapse
Affiliation(s)
- Ellen Caroline Feitoza Pires
- Department of Biotechnology, Molecular Biology Laboratory, Federal University of Tocantins, Gurupi 77410-570, Brazil; (E.C.F.P.); (R.W.d.S.A.)
- Department of Cell Biology, Institute of Biology, University of Brasília, Brasília 70910-900, Brazil
| | - Francini Pereira da Silva
- Institute of Health Sciences, Molecular Microbiology Laboratory, Feevale University, Novo Hamburgo 93525-075, Brazil (J.D.F.)
| | - Karoline Schallenberger
- Institute of Health Sciences, Molecular Microbiology Laboratory, Feevale University, Novo Hamburgo 93525-075, Brazil (J.D.F.)
| | - Bruna Saraiva Hermann
- Institute of Health Sciences, Molecular Microbiology Laboratory, Feevale University, Novo Hamburgo 93525-075, Brazil (J.D.F.)
| | - Larissa Mallmann
- Institute of Health Sciences, Molecular Microbiology Laboratory, Feevale University, Novo Hamburgo 93525-075, Brazil (J.D.F.)
| | - Wellington Souza Moura
- Department of Biotechnology Biodiversity and Graduate School of Biotechnology of Amazônia (Bionorte), Natural Products Laboratory, Federal University of Tocantins, Gurupi 77410-570, Brazil; (W.S.M.); (S.D.A.); (R.d.S.B.)
| | - Sergio Donizeti Ascêncio
- Department of Biotechnology Biodiversity and Graduate School of Biotechnology of Amazônia (Bionorte), Natural Products Laboratory, Federal University of Tocantins, Gurupi 77410-570, Brazil; (W.S.M.); (S.D.A.); (R.d.S.B.)
| | - Robson dos Santos Barbosa
- Department of Biotechnology Biodiversity and Graduate School of Biotechnology of Amazônia (Bionorte), Natural Products Laboratory, Federal University of Tocantins, Gurupi 77410-570, Brazil; (W.S.M.); (S.D.A.); (R.d.S.B.)
| | - Ilsamar Mendes Soares
- Department of Biotechnology Biodiversity and Graduate School of Biotechnology of Amazônia (Bionorte), Natural Products Laboratory, Federal University of Tocantins, Gurupi 77410-570, Brazil; (W.S.M.); (S.D.A.); (R.d.S.B.)
| | - Juliane Deise Fleck
- Institute of Health Sciences, Molecular Microbiology Laboratory, Feevale University, Novo Hamburgo 93525-075, Brazil (J.D.F.)
| | | | - Guy Smagghe
- Department of Plants and Crops, Ghent University, 9000 Ghent, Belgium
- Institute of Entomology, Guizhou University, Guiyang 550025, China
- Department of Biology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Bergmann Morais Ribeiro
- Department of Cell Biology, Institute of Biology, University of Brasília, Brasília 70910-900, Brazil
| | - Raimundo Wagner de Souza Aguiar
- Department of Biotechnology, Molecular Biology Laboratory, Federal University of Tocantins, Gurupi 77410-570, Brazil; (E.C.F.P.); (R.W.d.S.A.)
| |
Collapse
|
20
|
Mohd Azrin NA, Mohamad Ali MS, Raja Abd Rahman RNZ, Mohd Shariff F, Ahmad Kamarudin NH, Muhd Noor ND. Effect of cysteine mutation at Ca 2+ coordinating residues to the autolysis, folding and hydrophobicity of full length and mature Rand protease: molecular dynamics simulation and essential dynamics. J Biomol Struct Dyn 2024; 42:9018-9030. [PMID: 37608543 DOI: 10.1080/07391102.2023.2249105] [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: 04/10/2023] [Accepted: 08/12/2023] [Indexed: 08/24/2023]
Abstract
Rand protease is a serine protease that shared common characteristics with members of the MEROPS S8 subtilisin family. It is thermostable, highly stable in organic solvent and broad in specificity. Many structures of homologous protein solved by X-ray crystallography and NMR have been deposited to Protein Data Bank (PDB) which allowed this study to rely on structure prediction by deep learning to build three-dimensional (3D) structure of full length and mature Rand protease (flRP and mRP). In silico cysteine mutation to 7 predicted high affinity Ca2+ coordinating residues were introduced, and the mutants were subjected to molecular dynamics simulation to study its effect on flRP and mRP. MD simulation showed a marked increase in flexibility of the pro-peptide segment indicating the impact of single cysteine substitution at high affinity Ca2+ coordinating residues to autolysis of flRP. MD simulation for mRP reaffirmed the role of Ca2+ coordinating sites in providing stability to Rand protease. In addition, these residues also affect the autolysis, folding and hydrophobicity of RP. Essential dynamics observed large contribution of the first few eigenvectors of flRP, mRP and their high affinity Ca2+ coordinating residues mutants to the TMSF values which indicates that these values account for a large portion of the overall atomic fluctuations. These results have given a more comprehensive understanding on the role of cysteine substituted Ca2+ coordinating surface loop to the structure of flRP and mRP which are important in contributing to the structural stability of subtilisin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nur Aliyah Mohd Azrin
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
| | - Mohd Shukuri Mohamad Ali
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biochemistry, Universiti Putra Malaysia, Selangor, Malaysia
| | - Raja Noor Zaliha Raja Abd Rahman
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Microbiology, Universiti Putra Malaysia, Selangor, Malaysia
| | - Fairolniza Mohd Shariff
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Microbiology, Universiti Putra Malaysia, Selangor, Malaysia
| | - Nor Hafizah Ahmad Kamarudin
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
- Centre of Foundation Studies for Agricultural Science, Universiti Putra Malaysia, Selangor, Malaysia
| | - Noor Dina Muhd Noor
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biochemistry, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
21
|
Saberi M, Chikunova A, Ben Bdira F, Cramer-Blok A, Timmer M, Voskamp P, Ubbink M. Bimodal substrate binding in the active site of the glycosidase BcX. FEBS J 2024; 291:4222-4239. [PMID: 39185686 DOI: 10.1111/febs.17251] [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: 03/04/2024] [Revised: 05/02/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Bacillus circulans xylanase (BcX) from the glycoside hydrolase family 11 degrades xylan through a retaining, double-displacement mechanism. The enzyme is thought to hydrolyze glycosidic bonds in a processive manner and has a large, active site cleft, with six subsites allowing the binding of six xylose units. Such an active site architecture suggests that oligomeric xylose substrates can bind in multiple ways. In the crystal structure of the catalytically inactive variant BcX E78Q, the substrate xylotriose is observed in the active site, as well as bound to the known secondary binding site and a third site on the protein surface. Nuclear magnetic resonance (NMR) titrations with xylose oligomers of different lengths yield nonlinear chemical shift trajectories for active site nuclei resonances, indicative of multiple binding orientations for these substrates for which binding and dissociation are in fast exchange on the NMR timescale, exchanging on the micro- to millisecond timescale. Active site binding can be modeled with a 2 : 1 model with dissociation constants in the low and high millimolar range. Extensive mutagenesis of active site residues indicates that tight binding occurs in the glycon binding site and is stabilized by Trp9 and the thumb region. Mutations F125A and W71A lead to large structural rearrangements. Binding at the glycon site is sensed throughout the active site, whereas the weak binding mostly affects the aglycon site. The interactions with the two active site locations are largely independent of each other and of binding at the secondary binding site.
Collapse
Affiliation(s)
- Mahin Saberi
- Leiden Institute of Chemistry, Leiden University, The Netherlands
| | | | - Fredj Ben Bdira
- Leiden Institute of Chemistry, Leiden University, The Netherlands
| | | | - Monika Timmer
- Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Patrick Voskamp
- Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Marcellus Ubbink
- Leiden Institute of Chemistry, Leiden University, The Netherlands
| |
Collapse
|
22
|
Giovannuzzi S, Shyamal SS, Bhowmik R, Ray R, Manaithiya A, Carta F, Parrkila S, Aspatwar A, Supuran CT. Physiological modeling of the metaverse of the Mycobacterium tuberculosis β-CA inhibition mechanism. Comput Biol Med 2024; 181:109029. [PMID: 39173489 DOI: 10.1016/j.compbiomed.2024.109029] [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: 05/29/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
Tuberculosis (TB) is an infectious disease that primarily affects the lungs of humans and accounts for Mycobacterium tuberculosis (Mtb) bacteria as the etiologic agent. In this study, we introduce a computational framework designed to identify the important chemical features crucial for the effective inhibition of Mtb β-CAs. Through applying a mechanistic model, we elucidated the essential features pivotal for robust inhibition. Using this model, we engineered molecules that exhibit potent inhibitory activity and introduce relevant novel chemistry. The designed molecules were prioritized for synthesis based on their predicted pKi values via the QSAR (Quantitative Structure-Activity Relationship) model. All the rationally designed and synthesized compounds were evaluated in vitro against different carbonic anhydrase isoforms expressed from the pathogen Mtb; moreover, the off-target and widely human-expressed CA I and II were also evaluated. Among the reported derivatives, 2, 4, and 5 demonstrated the most valuable in vitro activity, resulting in promising candidates for the treatment of TB infection. All the synthesized molecules exhibited favorable pharmacokinetic and toxicological profiles based on in silico predictions. Docking analysis confirmed that the zinc-binding groups bind effectively into the catalytic triad of the Mtb β-Cas, supporting the in vitro outcomes with these binding interactions. Furthermore, molecules with good prediction accuracies according to previously established mechanistic and QSAR models were utilized to delve deeper into the realm of systems biology to understand their mechanism in combating tuberculotic pathogenesis. The results pointed to the key involvement of the compounds in modulating immune responses via NF-κβ1, SRC kinase, and TNF-α to modulate granuloma formation and clearance via T cells. This dual action, in which the pathogen's enzyme is inhibited while modulating the human immune machinery, represents a paradigm shift toward more effective and comprehensive treatment approaches for combating tuberculosis.
Collapse
Affiliation(s)
- Simone Giovannuzzi
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Sagar Singh Shyamal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ratul Bhowmik
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Rajarshi Ray
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ajay Manaithiya
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Fabrizio Carta
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Seppo Parrkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Claudiu T Supuran
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy.
| |
Collapse
|
23
|
Zhang T, Liu Y, Cao J, Jiang L, Wang P, Ren F, Yi H. Exploration of dynamic interaction between β-lactoglobulin and casein micelles during UHT milk process. Int J Biol Macromol 2024; 277:134367. [PMID: 39089562 DOI: 10.1016/j.ijbiomac.2024.134367] [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: 04/19/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The protein aggregation induced by UHT treatment shortens the shelf life of UHT milk. However, the mechanism of β-Lg induced casein micelle aggregation remains unclear. Herein, the dynamic interaction between β-Lg and casein micelles during UHT processing was investigated by experimental techniques and molecular dynamics simulations. Results showed that β-Lg decreased the stability of casein micelles, increased their size and zeta potential. Raman and FTIR spectra analysis suggested that hydrogen and disulfide bonds facilitated their interaction. Cryo-TEM showed that the formation of the casein micelle/β-Lg complex involved rigid binding, flexible linking, and severe cross-linking aggregation during UHT processing. SAXS and MST demonstrated β-Lg bound to κ-casein on micelle surfaces with a dissociation constant (Kd) of 3.84 ± 1.14 μm. Molecular docking and dynamic simulations identified the interacting amino acid residues and clarified that electrostatic and van der Waals forces drove the interaction. UHT treatment increased hydrogen bonds and decreased total binding energy. The non-covalent binding promoted the formation of disulfide bonds between β-Lg and casein micelles under heat treatment. Ultimately, it was concluded that non-covalent interaction and disulfide bonding resulted in casein micelle/β-Lg aggregates. These findings provided scientific insights into protein aggregation in UHT milk.
Collapse
Affiliation(s)
- Tai Zhang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Yisuo Liu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Jiayuan Cao
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Lu Jiang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Pengjie Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| | - Huaxi Yi
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| |
Collapse
|
24
|
Yadav H, Bakshi A, Anamika, Singh V, Paul P, Murugan NA, Maurya SK. Co-localization and co-expression of Olfml3 with Iba1 in brain of mice. J Neuroimmunol 2024; 394:578411. [PMID: 39079458 DOI: 10.1016/j.jneuroim.2024.578411] [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: 06/11/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/30/2024]
Abstract
Olfml3 is a microglia-specific protein whose role in neuroinflammation is elusive. In silico analysis was conducted to characterize the Olfml3 protein, followed by molecular docking and MD simulation to check possible interaction with Iba1. Further, expression and co-localization analysis was performed in the LPS-induced neuroinflammatory mice brains. Results suggest that Olfml3 physically interacts with Iba1. Olfml3 and Iba1 expression increases during neuroinflammation in mice brains. Olfml3 was observed to co-localize with Iba1, and the number of Olfml3 and Iba1 dual-positive cells increased in the brain of the neuroinflammatory mice model. Thus, Olfml3 could potentially participate in microglia functions by interacting with Iba1.
Collapse
Affiliation(s)
- Himanshi Yadav
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Amrita Bakshi
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Anamika
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Vishal Singh
- Electron Microscope Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Prateek Paul
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Delhi, India
| | - N Arul Murugan
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Delhi, India
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India.
| |
Collapse
|
25
|
de Carvalho GA, Tambwe PM, Nascimento LRC, Campos BKP, Chiareli RA, Junior GPN, Menegatti R, Gomez RS, Pinto MCX. In silico evidence of bitopertin's broad interactions within the SLC6 transporter family. J Pharm Pharmacol 2024; 76:1199-1211. [PMID: 38982944 DOI: 10.1093/jpp/rgae051] [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: 01/12/2024] [Accepted: 04/16/2024] [Indexed: 07/11/2024]
Abstract
The Glycine Transporter Type 1 (GlyT1) significantly impacts central nervous system functions, influencing glycinergic and glutamatergic neurotransmission. Bitopertin, the first GlyT1 inhibitor in clinical trials, was developed for schizophrenia treatment but showed limited efficacy. Despite this, bitopertin's repositioning could advance treating various pathologies. This study aims to understand bitopertin's mechanism of action using computational methods, exploring off-target effects, and providing a comprehensive pharmacological profile. Similarity Ensemble Approach (SEA) and SwissTargetPrediction initially predicted targets, followed by molecular modeling on SWISS-MODEL and GalaxyWeb servers. Binding sites were identified using PrankWeb, and molecular docking was performed with DockThor and GOLD software. Molecular dynamics analyses were conducted on the Visual Dynamics platform. Reverse screening on SEA and SwissTargetPrediction identified GlyT1 (SLC6A9), GlyT2 (SLC6A5), PROT (SLC6A7), and DAT (SLC6A3) as potential bitopertin targets. Homology modeling on SwissModel generated high-resolution models, optimized further on GalaxyWeb. PrankWeb identified similar binding sites in GlyT1, GlyT2, PROT, and DAT, indicating potential interaction. Docking studies suggested bitopertin's interaction with GlyT1 and proximity to GlyT2 and PROT. Molecular dynamics confirmed docking results, highlighting bitopertin's target stability beyond GlyT1. The study concludes that bitopertin potentially interacts with multiple SLC6 family targets, indicating a broader pharmacological property.
Collapse
Affiliation(s)
- Gustavo Almeida de Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| | - Paul Magogo Tambwe
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| | - Lucas Rodrigues Couto Nascimento
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| | - Bruna Kelly Pedrosa Campos
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| | - Raphaela Almeida Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| | - Guilhermino Pereira Nunes Junior
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| | - Ricardo Menegatti
- Faculdade de Farmácia, Universidade Federal de Goiás, Rua 240, Setor Leste Universitário, 74605170 - Goiânia, GO, Brazil
| | - Renato Santiago Gomez
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190, 30130-100, Belo Horizonte-MG, Brazil
| | - Mauro Cunha Xavier Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, CEP 74690-900, Goiânia-GO, Brazil
| |
Collapse
|
26
|
Gorrab A, Ouertani R, Hammami K, Souii A, Kallel F, Masmoudi AS, Cherif A, Neifar M. In silico and experimental characterization of a new polyextremophilic subtilisin-like protease from Microbacterium metallidurans and its application as a laundry detergent additive. 3 Biotech 2024; 14:200. [PMID: 39144069 PMCID: PMC11319565 DOI: 10.1007/s13205-024-04043-1] [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: 06/27/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
Considering the current growing interest in new and improved enzymes for use in a variety of applications, the present study aimed to characterize a novel detergent-stable serine alkaline protease from the extremophilic actinobacterium Microbacterium metallidurans TL13 (MmSP) using a combined in silico and experimental approach. The MmSP showed a close phylogenetic relationship with high molecular weight S8 peptidases of Microbacterium species. Moreover, its physical and chemical parameters computed using Expasy's ProtParam tool revealed that MmSP is hydrophilic, halophilic and thermo-alkali stable. 3D structure modelling and functional prediction of TL13 serine protease resulted in the detection of five characteristic domains: [catalytic subtilase domain, fibronectin (Fn) type-III domain, peptidase inhibitor I9, protease-associated (PA) domain and bacterial Ig-like domain (group 3)], as well as the three amino acid residues [aspartate (D182), histidine (H272) and serine (S604)] in the catalytic subtilase domain. The extremophilic strain TL13 was tested for protease production using agricultural wastes/by-products as carbon substrates. Maximum enzyme activity (390 U/gds) was obtained at 8th day fermentation on potato peel medium. Extracellular extract was concentrated and partially purified using ammonium sulfate precipitation methodology (1.58 folds purification fold). The optimal pH, temperature and salinity of MmSP were 9, 60 °C and 1 M NaCl, respectively. The MmSP protease showed broad pH stability, thermal stability, salt tolerance and detergent compatibility. In order to achieve the maximum stain removal efficacy by the TL 13 serine protease, the operation conditions were optimized using a Box-Behnken Design (BBD) with four variables, namely, time (15-75 min), temperature (30-60 °C), MmSP enzyme concentration (5-10 U/mL) and pH (7-11). The maximum stain removal yield (95 ± 4%) obtained under the optimal enzymatic operation conditions (treatment with 7.5 U/mL of MmSP during 30 min at 32 °C and pH9) was in good agreement with the value predicted by the regression model (98 ± %), which prove the validity of the fitted model. In conclusion, MmSP appears to be a good candidate for industrial applications, particularly in laundry detergent formulations, due to its high hydrophilicity, alkali-halo-stability, detergent compatibility and stain removal efficiency.
Collapse
Affiliation(s)
- Afwa Gorrab
- Laboratory BVBGR-LR11ES31, Institute of Biotechnology of Sidi Thabet, Biotechpole Sidi Thabet, 2020 Ariana, Tunisia
| | - Rania Ouertani
- Laboratory BVBGR-LR11ES31, Institute of Biotechnology of Sidi Thabet, Biotechpole Sidi Thabet, 2020 Ariana, Tunisia
| | - Khouloud Hammami
- Laboratory BVBGR-LR11ES31, Institute of Biotechnology of Sidi Thabet, Biotechpole Sidi Thabet, 2020 Ariana, Tunisia
| | - Amal Souii
- Laboratory BVBGR-LR11ES31, Institute of Biotechnology of Sidi Thabet, Biotechpole Sidi Thabet, 2020 Ariana, Tunisia
| | - Fatma Kallel
- Laboratory of Plant Improvement and Valorization of Agro-resources (APVA-LR16ES20), ENIS, University of Sfax, 3030 Sfax, Tunisia
| | - Ahmed Slaheddine Masmoudi
- Laboratory BVBGR-LR11ES31, Institute of Biotechnology of Sidi Thabet, Biotechpole Sidi Thabet, 2020 Ariana, Tunisia
| | - Ameur Cherif
- Laboratory BVBGR-LR11ES31, Institute of Biotechnology of Sidi Thabet, Biotechpole Sidi Thabet, 2020 Ariana, Tunisia
| | - Mohamed Neifar
- Laboratory of Plant Improvement and Valorization of Agro-resources (APVA-LR16ES20), ENIS, University of Sfax, 3030 Sfax, Tunisia
- Common Services Unit “Bioreactor Coupled with an Ultrafilter”, ENIS, University of Sfax, 3030 Sfax, Tunisia
| |
Collapse
|
27
|
Schulz L, Ung KL, Koutnik-Abele S, Stokes DL, Pedersen BP, Hammes UZ. Transport of herbicides by PIN-FORMED auxin transporters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610046. [PMID: 39257797 PMCID: PMC11383987 DOI: 10.1101/2024.08.29.610046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Auxins are a group of phytohormones that control plant growth and development 1. Their crucial role in plant physiology has inspired development of potent synthetic auxins that can be used as herbicides 2. Phenoxyacetic acid derivatives are a widely used group of auxin herbicides in agriculture and research. Despite their prevalence, the identity of the transporters required for distribution of these herbicides in plants is both poorly understood and the subject of controversial debate 3,4. Here we show that PIN-FORMED auxin transporters transport a range of phenoxyacetic acid herbicides across the membrane and we characterize the molecular determinants of this process using a variety of different substrates as well as protein mutagenesis to control substrate specificity. Finally, we present Cryo-EM structures of Arabidopsis thaliana PIN8 with 2,4-dichlorophenoxyacetic acid (2,4-D) or 4-chlorophenoxyacetic acid (4-CPA) bound. These structures represent five key states from the transport cycle, allowing us to describe conformational changes associated with substrate binding and transport across the membrane. Overall, our results reveal that phenoxyacetic acid herbicides use the same export machinery as endogenous auxins and exemplify how transporter binding sites undergo transformations that dictate substrate specificity. These results enable development of novel synthetic auxins and for guiding precision breeding of herbicide resistant crop plants.
Collapse
Affiliation(s)
- Lukas Schulz
- Plant Systems Biology, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Kien Lam Ung
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Sarah Koutnik-Abele
- Plant Systems Biology, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- Institute for Advanced Study, Technical University of Munich, D-85748 Garching, Germany
| | - David L Stokes
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York 10016, United States of America
| | - Bjørn Panyella Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
- Institute for Advanced Study, Technical University of Munich, D-85748 Garching, Germany
| | - Ulrich Z. Hammes
- Plant Systems Biology, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
28
|
Silva-Rodrigues G, de Castro IM, Borges PHG, Suzukawa HT, de Souza JM, Bartolomeu-Gonçalves G, Pelisson M, Medeiros CIS, Bispo MDLF, de Almeida RSC, Ishida K, Tavares ER, Yamauchi LM, Yamada-Ogatta SF. Geraniol Potentiates the Effect of Fluconazole against Planktonic and Sessile Cells of Azole-Resistant Candida tropicalis: In Vitro and In Vivo Analyses. Pharmaceutics 2024; 16:1053. [PMID: 39204397 PMCID: PMC11360560 DOI: 10.3390/pharmaceutics16081053] [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: 06/27/2024] [Revised: 07/24/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Candida tropicalis is regarded as an opportunistic pathogen, causing diseases ranging from superficial infections to life-threatening disseminated infections. The ability of this yeast to form biofilms and develop resistance to antifungals represents a significant therapeutic challenge. Herein, the effect of geraniol (GER), alone and combined with fluconazole (FLZ), was evaluated in the planktonic and sessile cells of azole-resistant C. tropicalis. GER showed a time-dependent fungicidal effect on the planktonic cells, impairing the cell membrane integrity. Additionally, GER inhibited the rhodamine 6G efflux, and the molecular docking analyzes supported the binding affinity of GER to the C. tropicalis Cdr1 protein. GER exhibited a synergism with FLZ against the planktonic and sessile cells, inhibiting the adhesion of the yeast cells and the viability of the 48-h biofilms formed on abiotic surfaces. C. tropicalis biofilms treated with GER, alone or combined with FLZ, displayed morphological and ultrastructural alterations, including a decrease in the stacking layers and the presence of wilted cells. Moreover, neither GER alone nor combined with FLZ caused toxicity, and both treatments prolonged the survival of the Galleria mellonella larvae infected with azole-resistant C. tropicalis. These findings indicate that the combination of GER and FLZ may be a promising strategy to control azole-resistant C. tropicalis infections.
Collapse
Affiliation(s)
- Gislaine Silva-Rodrigues
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Isabela Madeira de Castro
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Paulo Henrique Guilherme Borges
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Helena Tiemi Suzukawa
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Joyce Marinho de Souza
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Guilherme Bartolomeu-Gonçalves
- Postgraduate Program in Clinical and Laboratory Pathophysiology, Department of Pathology, Clinical and Toxicological Analysis, State University of Londrina, Londrina 86038-350, Brazil
| | - Marsileni Pelisson
- Postgraduate Program in Clinical and Laboratory Pathophysiology, Department of Pathology, Clinical and Toxicological Analysis, State University of Londrina, Londrina 86038-350, Brazil
| | | | - Marcelle de Lima Ferreira Bispo
- Synthesis of Medicinal Molecules Laboratory, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil;
| | - Ricardo Sérgio Couto de Almeida
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Kelly Ishida
- Laboratory of Antifungal Chemotherapy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil;
| | - Eliandro Reis Tavares
- Department of Medicine, Pontifical Catholic University of Paraná, Campus Londrina, Londrina 86067-000, Brazil;
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil
| | - Lucy Megumi Yamauchi
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil
| | - Sueli Fumie Yamada-Ogatta
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
- Postgraduate Program in Clinical and Laboratory Pathophysiology, Department of Pathology, Clinical and Toxicological Analysis, State University of Londrina, Londrina 86038-350, Brazil
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil
| |
Collapse
|
29
|
Loaiza-Cano V, Hernández-Mira E, Pastrana-Restrepo M, Galeano E, Pardo-Rodriguez D, Martinez-Gutierrez M. The Mechanism of Action of L-Tyrosine Derivatives against Chikungunya Virus Infection In Vitro Depends on Structural Changes. Int J Mol Sci 2024; 25:7972. [PMID: 39063216 PMCID: PMC11277544 DOI: 10.3390/ijms25147972] [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: 06/20/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Although the disease caused by chikungunya virus (CHIKV) is of great interest to public health organizations around the world, there are still no authorized antivirals for its treatment. Previously, dihalogenated anti-CHIKV compounds derived from L-tyrosine (dH-Y) were identified as being effective against in vitro infection by this virus, so the objective of this study was to determine the mechanisms of its antiviral action. Six dH-Y compounds (C1 to C6) dihalogenated with bromine or chlorine and modified in their amino groups were evaluated by different in vitro antiviral strategies and in silico tools. When the cells were exposed before infection, all compounds decreased the expression of viral proteins; only C4, C5 and C6 inhibited the genome; and C1, C2 and C3 inhibited infectious viral particles (IVPs). Furthermore, C1 and C3 reduce adhesion, while C2 and C3 reduce internalization, which could be related to the in silico interaction with the fusion peptide of the E1 viral protein. Only C3, C4, C5 and C6 inhibited IVPs when the cells were exposed after infection, and their effect occurred in late stages after viral translation and replication, such as assembly, and not during budding. In summary, the structural changes of these compounds determine their mechanism of action. Additionally, C3 was the only compound that inhibited CHIKV infection at different stages of the replicative cycle, making it a compound of interest for conversion as a potential drug.
Collapse
Affiliation(s)
- Vanessa Loaiza-Cano
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia; (V.L.-C.); (E.H.-M.)
| | - Estiven Hernández-Mira
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia; (V.L.-C.); (E.H.-M.)
| | - Manuel Pastrana-Restrepo
- Grupo de Investigación en Productos Naturales Marinos, Universidad de Antioquia, Medellin 050010, Colombia; (M.P.-R.); (E.G.)
| | - Elkin Galeano
- Grupo de Investigación en Productos Naturales Marinos, Universidad de Antioquia, Medellin 050010, Colombia; (M.P.-R.); (E.G.)
| | - Daniel Pardo-Rodriguez
- Metabolomics Core Facility—MetCore, Vice-Presidency for Research, Universidad de los Andes, Bogota 111711, Colombia;
| | - Marlen Martinez-Gutierrez
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia; (V.L.-C.); (E.H.-M.)
- Grupo de Investigación en Microbiología Básica y Aplicada (MICROBA), Escuela de Microbiología, Universidad de Antioquia, Medellin 050010, Colombia
| |
Collapse
|
30
|
Zubatyuk R, Biczysko M, Ranasinghe K, Moriarty NW, Gokcan H, Kruse H, Poon BK, Adams PD, Waller MP, Roitberg AE, Isayev O, Afonine PV. AQuaRef: Machine learning accelerated quantum refinement of protein structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.21.604493. [PMID: 39071315 PMCID: PMC11275739 DOI: 10.1101/2024.07.21.604493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Cryo-EM and X-ray crystallography provide crucial experimental data for obtaining atomic-detail models of biomacromolecules. Refining these models relies on library-based stereochemical restraints, which, in addition to being limited to known chemical entities, do not include meaningful noncovalent interactions relying solely on nonbonded repulsions. Quantum mechanical (QM) calculations could alleviate these issues but are too expensive for large molecules. We present a novel AI-enabled Quantum Refinement (AQuaRef) based on AIMNet2 neural network potential mimicking QM at substantially lower computational costs. By refining 41 cryo-EM and 30 X-ray structures, we show that this approach yields atomic models with superior geometric quality compared to standard techniques, while maintaining an equal or better fit to experimental data.
Collapse
Affiliation(s)
- Roman Zubatyuk
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Malgorzata Biczysko
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland
| | | | - Nigel W. Moriarty
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
| | - Hatice Gokcan
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | - Billy K. Poon
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
| | - Paul D. Adams
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720, USA
| | | | - Adrian E. Roitberg
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Olexandr Isayev
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Pavel V. Afonine
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
| |
Collapse
|
31
|
Chen J, Zehr EA, Gruschus JM, Szyk A, Liu Y, Tanner ME, Tjandra N, Roll-Mecak A. Tubulin code eraser CCP5 binds branch glutamates by substrate deformation. Nature 2024; 631:905-912. [PMID: 39020174 DOI: 10.1038/s41586-024-07699-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 06/11/2024] [Indexed: 07/19/2024]
Abstract
Microtubule function is modulated by the tubulin code, diverse posttranslational modifications that are altered dynamically by writer and eraser enzymes1. Glutamylation-the addition of branched (isopeptide-linked) glutamate chains-is the most evolutionarily widespread tubulin modification2. It is introduced by tubulin tyrosine ligase-like enzymes and erased by carboxypeptidases of the cytosolic carboxypeptidase (CCP) family1. Glutamylation homeostasis, achieved through the balance of writers and erasers, is critical for normal cell function3-9, and mutations in CCPs lead to human disease10-13. Here we report cryo-electron microscopy structures of the glutamylation eraser CCP5 in complex with the microtubule, and X-ray structures in complex with transition-state analogues. Combined with NMR analysis, these analyses show that CCP5 deforms the tubulin main chain into a unique turn that enables lock-and-key recognition of the branch glutamate in a cationic pocket that is unique to CCP family proteins. CCP5 binding of the sequences flanking the branch point primarily through peptide backbone atoms enables processing of diverse tubulin isotypes and non-tubulin substrates. Unexpectedly, CCP5 exhibits inefficient processing of an abundant β-tubulin isotype in the brain. This work provides an atomistic view into glutamate branch recognition and resolution, and sheds light on homeostasis of the tubulin glutamylation syntax.
Collapse
Affiliation(s)
- Jiayi Chen
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Elena A Zehr
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - James M Gruschus
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Agnieszka Szyk
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Yanjie Liu
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin E Tanner
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nico Tjandra
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
32
|
Azmal M, Hossen MS, Shohan MNH, Taqui R, Malik A, Ghosh A. A computational approach to identify phytochemicals as potential inhibitor of acetylcholinesterase: Molecular docking, ADME profiling and molecular dynamics simulations. PLoS One 2024; 19:e0304490. [PMID: 38833492 PMCID: PMC11149856 DOI: 10.1371/journal.pone.0304490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/10/2024] [Indexed: 06/06/2024] Open
Abstract
Inhibition of acetylcholinesterase (AChE) is a crucial target in the treatment of Alzheimer's disease (AD). Common anti-acetylcholinesterase drugs such as Galantamine, Rivastigmine, Donepezil, and Tacrine have significant inhibition potential. Due to side effects and safety concerns, we aimed to investigate a wide range of phytochemicals and structural analogues of these compounds. Compounds similar to the established drugs, and phytochemicals were investigated as potential inhibitors for AChE in treating AD. A total of 2,270 compound libraries were generated for further analysis. Initial virtual screening was performed using Pyrx software, resulting in 638 molecules showing higher binding affinities compared to positive controls Tacrine (-9.0 kcal/mol), Donepezil (-7.3 kcal/mol), Galantamine (-8.3 kcal/mol), and Rivastigmine (-6.4 kcal/mol). Subsequently, ADME properties were assessed, including blood-brain barrier permeability and Lipinski's rule of five violations, leading to 88 compounds passing the ADME analysis. Among the rivastigmine analogous, [3-(1-methylpiperidin-2-yl)phenyl] N,N-diethylcarbamate showed interaction with Tyr123, Tyr336, Tyr340, Phe337, Trp285 residues of AChE. Tacrine similar compounds, such as 4-amino-2-styrylquinoline, exhibited bindings with Tyr123, Phe337, Tyr336, Trp285, Trp85, Gly119, and Gly120 residues. A phytocompound (bisdemethoxycurcumin) showed interaction with Trp285, Tyr340, Trp85, Tyr71, and His446 residues of AChE with favourable binding. These findings underscore the potential of these compounds as novel inhibitors of AChE, offering insights into alternative therapeutic avenues for AD. A 100ns simulation analysis confirmed the stability of protein-ligand complex based on the RMSD, RMSF, ligand properties, PCA, DCCM and MMGBS parameters. The investigation suggested 3 ligands as a potent inhibitor of AChE which are [3-(1-methylpiperidin-2-yl)phenyl] N,N-diethylcarbamate, 4-Amino-2-styrylquinoline and bisdemethoxycurcumin. Furthermore, investigation, including in-vitro and in-vivo studies, is needed to validate the efficacy, safety profiles, and therapeutic potential of these compounds for AD treatment.
Collapse
Affiliation(s)
- Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Sahadot Hossen
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Naimul Haque Shohan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Rashid Taqui
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Abbeha Malik
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Punjab, Pakistan
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
33
|
Sharma G, Singh V, Raheja Y, Chadha BS. Unlocking the potential of feruloyl esterase from Myceliophthora verrucosa: a key player in efficient conversion of biorefinery-relevant pretreated rice straw. 3 Biotech 2024; 14:168. [PMID: 38828098 PMCID: PMC11139844 DOI: 10.1007/s13205-024-04013-7] [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: 02/19/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The lignocellulolytic accessory enzyme, Feruloyl esterase C (FE_5DR), encoded in the genome of thermotolerant Myceliophthora verrucosa was successfully cloned and heterologously expressed in Pichia pastoris. The expressed FE_5DR was purified using UNOsphere™ Q anion exchange chromatography column, exhibiting a homogeneous band of ~ 39 kDa. Its optimum temperature was determined to be 60 °C, with an optimal pH of 6.0. Additionally, the enzyme activity of FE_5DR was significantly enhanced by preincubation in a buffer containing Mg2+, Cu2+ and Ca2 metal ions. Enzyme kinetic parameters, computed from double reciprocal Lineweaver-Burk plots, yielded observed Vmax and Km values of 0.758 U/mg and 0.439 mM, respectively. Furthermore, the potential of custom-made cocktails comprising FE_5DR and benchmark cellulase derived from the developed mutant strain of Aspergillus allahabadii MAN 40, as well as the biorefinery-relevant lignocellulolytic enzyme Cellic CTec 3, resulted in improved saccharification of unwashed acid pretreated (UWAP) rice straw slurry and mild alkali deacetylated (MAD) rice straw when compared to benchmark MAN 40 and Cellic CTec 3. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04013-7.
Collapse
Affiliation(s)
- Gaurav Sharma
- Department of Microbiology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Varinder Singh
- Department of Microbiology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Yashika Raheja
- Department of Microbiology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | | |
Collapse
|
34
|
Gonzalez-Llerena JL, Espinosa-Rodriguez BA, Treviño-Almaguer D, Mendez-Lopez LF, Carranza-Rosales P, Gonzalez-Barranco P, Guzman-Delgado NE, Romo-Mancillas A, Balderas-Renteria I. Cordycepin Triphosphate as a Potential Modulator of Cellular Plasticity in Cancer via cAMP-Dependent Pathways: An In Silico Approach. Int J Mol Sci 2024; 25:5692. [PMID: 38891880 PMCID: PMC11171877 DOI: 10.3390/ijms25115692] [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: 04/05/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Cordycepin, or 3'-deoxyadenosine, is an adenosine analog with a broad spectrum of biological activity. The key structural difference between cordycepin and adenosine lies in the absence of a hydroxyl group at the 3' position of the ribose ring. Upon administration, cordycepin can undergo an enzymatic transformation in specific tissues, forming cordycepin triphosphate. In this study, we conducted a comprehensive analysis of the structural features of cordycepin and its derivatives, contrasting them with endogenous purine-based metabolites using chemoinformatics and bioinformatics tools in addition to molecular dynamics simulations. We tested the hypothesis that cordycepin triphosphate could bind to the active site of the adenylate cyclase enzyme. The outcomes of our molecular dynamics simulations revealed scores that are comparable to, and superior to, those of adenosine triphosphate (ATP), the endogenous ligand. This interaction could reduce the production of cyclic adenosine monophosphate (cAMP) by acting as a pseudo-ATP that lacks a hydroxyl group at the 3' position, essential to carry out nucleotide cyclization. We discuss the implications in the context of the plasticity of cancer and other cells within the tumor microenvironment, such as cancer-associated fibroblast, endothelial, and immune cells. This interaction could awaken antitumor immunity by preventing phenotypic changes in the immune cells driven by sustained cAMP signaling. The last could be an unreported molecular mechanism that helps to explain more details about cordycepin's mechanism of action.
Collapse
Affiliation(s)
- Jose Luis Gonzalez-Llerena
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
- Center for Research on Nutrition and Public Health, School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 66460, Mexico;
| | - Bryan Alejandro Espinosa-Rodriguez
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Daniela Treviño-Almaguer
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Luis Fernando Mendez-Lopez
- Center for Research on Nutrition and Public Health, School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 66460, Mexico;
| | - Pilar Carranza-Rosales
- Laboratory of Cell Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Mexico;
| | - Patricia Gonzalez-Barranco
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Nancy Elena Guzman-Delgado
- Health Research Division, High Specialty Medical Unit, Cardiology Hospital N. 34. Mexican Social Security Institute, Monterrey 64360, Mexico;
| | - Antonio Romo-Mancillas
- Computer Aided Drug Design and Synthesis Group, School of Chemistry, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | - Isaias Balderas-Renteria
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| |
Collapse
|
35
|
Frain KM, Dedic E, Nel L, Bohush A, Olesen E, Thaysen K, Wüstner D, Stokes DL, Pedersen BP. Conformational changes in the Niemann-Pick type C1 protein NCR1 drive sterol translocation. Proc Natl Acad Sci U S A 2024; 121:e2315575121. [PMID: 38568972 PMCID: PMC11009665 DOI: 10.1073/pnas.2315575121] [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: 09/08/2023] [Accepted: 02/22/2024] [Indexed: 04/05/2024] Open
Abstract
The membrane protein Niemann-Pick type C1 (NPC1, named NCR1 in yeast) is central to sterol homeostasis in eukaryotes. Saccharomyces cerevisiae NCR1 is localized to the vacuolar membrane, where it is suggested to carry sterols across the protective glycocalyx and deposit them into the vacuolar membrane. However, documentation of a vacuolar glycocalyx in fungi is lacking, and the mechanism for sterol translocation has remained unclear. Here, we provide evidence supporting the presence of a glycocalyx in isolated S. cerevisiae vacuoles and report four cryo-EM structures of NCR1 in two distinct conformations, named tense and relaxed. These two conformations illustrate the movement of sterols through a tunnel formed by the luminal domains, thus bypassing the barrier presented by the glycocalyx. Based on these structures and on comparison with other members of the Resistance-Nodulation-Division (RND) superfamily, we propose a transport model that links changes in the luminal domains with a cycle of protonation and deprotonation within the transmembrane region of the protein. Our model suggests that NPC proteins work by a generalized RND mechanism where the proton motive force drives conformational changes in the transmembrane domains that are allosterically coupled to luminal/extracellular domains to promote sterol transport.
Collapse
Affiliation(s)
- Kelly M. Frain
- Department of Molecular Biology and Genetics, Aarhus University, AarhusC 8000, Denmark
| | - Emil Dedic
- Department of Molecular Biology and Genetics, Aarhus University, AarhusC 8000, Denmark
| | - Lynette Nel
- Department of Molecular Biology and Genetics, Aarhus University, AarhusC 8000, Denmark
| | - Anastasiia Bohush
- Department of Molecular Biology and Genetics, Aarhus University, AarhusC 8000, Denmark
- Department of Molecular Biology and Genetics, Aarhus Institute of Advanced Studies, Aarhus University, AarhusC 8000, Denmark
| | - Esben Olesen
- Department of Molecular Biology and Genetics, Aarhus University, AarhusC 8000, Denmark
| | - Katja Thaysen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, OdenseM 5230, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, OdenseM 5230, Denmark
| | - David L. Stokes
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY10016
| | | |
Collapse
|
36
|
Chikunova A, Manley MP, Heijjer CN, Drenth CS, Cramer-Blok AJ, Ahmad MUD, Perrakis A, Ubbink M. Conserved proline residues prevent dimerization and aggregation in the β-lactamase BlaC. Protein Sci 2024; 33:e4972. [PMID: 38533527 DOI: 10.1002/pro.4972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
Evolution leads to conservation of amino acid residues in protein families. Conserved proline residues are usually considered to ensure the correct folding and to stabilize the three-dimensional structure. Surprisingly, proline residues that are highly conserved in class A β-lactamases were found to tolerate various substitutions without large losses in enzyme activity. We investigated the roles of three conserved prolines at positions 107, 226, and 258 in the β-lactamase BlaC from Mycobacterium tuberculosis and found that mutations can lead to dimerization of the enzyme and an overall less stable protein that is prone to aggregate over time. For the variant Pro107Thr, the crystal structure shows dimer formation resembling domain swapping. It is concluded that the proline substitutions loosen the structure, enhancing multimerization. Even though the enzyme does not lose its properties without the conserved proline residues, the prolines ensure the long-term structural integrity of the enzyme.
Collapse
Affiliation(s)
- A Chikunova
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - M P Manley
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - C N Heijjer
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - C S Drenth
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - A J Cramer-Blok
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - M Ud Din Ahmad
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Perrakis
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Ubbink
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Department of Infectious Diseases, Imperial College, London, UK
- Zocdoc, New York City, New York, USA
- ZoBio BV, Leiden, The Netherlands
| |
Collapse
|
37
|
Yu F, Wu X, Chen W, Yan F, Li W. Computer-assisted discovery and evaluation of potential ribosomal protein S6 kinase beta 2 inhibitors. Comput Biol Med 2024; 172:108204. [PMID: 38484695 DOI: 10.1016/j.compbiomed.2024.108204] [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: 11/23/2023] [Revised: 02/11/2024] [Accepted: 02/19/2024] [Indexed: 03/26/2024]
Abstract
S6K2 is an important protein in mTOR signaling pathway and cancer. To identify potential S6K2 inhibitors for mTOR pathway treatment, a virtual screening of 1,575,957 active molecules was performed using PLANET, AutoDock GPU, and AutoDock Vina, with their classification abilities compared. The MM/PB(GB)SA method was used to identify four compounds with the strongest binding energies. These compounds were further investigated using molecular dynamics (MD) simulations to understand the properties of the S6K2/ligand complex. Due to a lack of available 3D structures of S6K2, OmegaFold served as a reliable 3D predictive model with higher evaluation scores in SAVES v6.0 than AlphaFold, AlphaFold2, and RoseTTAFold2. The 150 ns MD simulation revealed that the S6K2 structure in aqueous solvation experienced compression during conformational relaxation and encountered potential energy traps of about 19.6 kJ mol-1. The virtual screening results indicated that Lys75 and Lys99 in S6K2 are key binding sites in the binding cavity. Additionally, MD simulations revealed that the ligands remained attached to the activation cavity of S6K2. Among the compounds, compound 1 induced restrictive dissociation of S6K2 in the presence of a flexible region, compound 8 achieved strong stability through hydrogen bonding with Lys99, compound 9 caused S6K2 tightening, and the binding of compound 16 was heavily influenced by hydrophobic interactions. This study suggests that these four potential inhibitors with different mechanisms of action could provide potential therapeutic options.
Collapse
Affiliation(s)
- Fangyi Yu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xiaochuan Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - WeiSong Chen
- Department of Respiratory Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Fugui Yan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
38
|
Samudrala M, Dhaveji S, Savsani K, Dakshanamurthy S. AutoEpiCollect, a Novel Machine Learning-Based GUI Software for Vaccine Design: Application to Pan-Cancer Vaccine Design Targeting PIK3CA Neoantigens. Bioengineering (Basel) 2024; 11:322. [PMID: 38671743 PMCID: PMC11048108 DOI: 10.3390/bioengineering11040322] [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: 02/27/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Previous epitope-based cancer vaccines have focused on analyzing a limited number of mutated epitopes and clinical variables preliminarily to experimental trials. As a result, relatively few positive clinical outcomes have been observed in epitope-based cancer vaccines. Further efforts are required to diversify the selection of mutated epitopes tailored to cancers with different genetic signatures. To address this, we developed the first version of AutoEpiCollect, a user-friendly GUI software, capable of generating safe and immunogenic epitopes from missense mutations in any oncogene of interest. This software incorporates a novel, machine learning-driven epitope ranking method, leveraging a probabilistic logistic regression model that is trained on experimental T-cell assay data. Users can freely download AutoEpiCollectGUI with its user guide for installing and running the software on GitHub. We used AutoEpiCollect to design a pan-cancer vaccine targeting missense mutations found in the proto-oncogene PIK3CA, which encodes the p110ɑ catalytic subunit of the PI3K kinase protein. We selected PIK3CA as our gene target due to its widespread prevalence as an oncokinase across various cancer types and its lack of presence as a gene target in clinical trials. After entering 49 distinct point mutations into AutoEpiCollect, we acquired 361 MHC Class I epitope/HLA pairs and 219 MHC Class II epitope/HLA pairs. From the 49 input point mutations, we identified MHC Class I epitopes targeting 34 of these mutations and MHC Class II epitopes targeting 11 mutations. Furthermore, to assess the potential impact of our pan-cancer vaccine, we employed PCOptim and PCOptim-CD to streamline our epitope list and attain optimized vaccine population coverage. We achieved a world population coverage of 98.09% for MHC Class I data and 81.81% for MHC Class II data. We used three of our predicted immunogenic epitopes to further construct 3D models of peptide-HLA and peptide-HLA-TCR complexes to analyze the epitope binding potential and TCR interactions. Future studies could aim to validate AutoEpiCollect's vaccine design in murine models affected by PIK3CA-mutated or other mutated tumor cells located in various tissue types. AutoEpiCollect streamlines the preclinical vaccine development process, saving time for thorough testing of vaccinations in experimental trials.
Collapse
Affiliation(s)
- Madhav Samudrala
- College of Arts and Sciences, The University of Virginia, Charlottesville, VA 22903, USA
| | | | - Kush Savsani
- College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA 22043, USA
| | - Sivanesan Dakshanamurthy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| |
Collapse
|
39
|
Hussin A, Nathan S, Shahidan MA, Nor Rahim MY, Zainun MY, Khairuddin NAN, Ibrahim N. Identification and mechanism determination of the efflux pump subunit amrB gene mutations linked to gentamicin susceptibility in clinical Burkholderia pseudomallei from Malaysian Borneo. Mol Genet Genomics 2024; 299:12. [PMID: 38381232 DOI: 10.1007/s00438-024-02105-w] [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: 07/18/2023] [Accepted: 12/29/2023] [Indexed: 02/22/2024]
Abstract
The bacterium Burkholderia pseudomallei is typically resistant to gentamicin but rare susceptible strains have been isolated in certain regions, such as Thailand and Sarawak, Malaysia. Recently, several amino acid substitutions have been reported in the amrB gene (a subunit of the amrAB-oprA efflux pump gene) that confer gentamicin susceptibility. However, information regarding the mechanism of the substitutions conferring the susceptibility is lacking. To understand the mechanism of amino acid substitution that confers susceptibility, this study identifies the corresponding mutations in clinical gentamicin-susceptible B. pseudomallei isolates from the Malaysian Borneo (n = 46; Sarawak: 5; Sabah: 41). Three phenotypically confirmed gentamicin-susceptible (GENs) strains from Sarawak, Malaysia, were screened for mutations in the amrB gene using gene sequences of gentamicin-resistant (GENr) strains (QEH 56, QEH 57, QEH20, and QEH26) and publicly available sequences (AF072887.1 and BX571965.1) as the comparator. The effect of missense mutations on the stability of the AmrB protein was determined by calculating the average energy change value (ΔΔG). Mutagenesis analysis identified a polymorphism-associated mutation, g.1056 T > G, a possible susceptible-associated in-frame deletion, Delta V412, and a previously confirmed susceptible-associated amino acid substitution, T368R, in each of the three GENs isolates. The contribution of Delta V412 needs further confirmation by experimental mutagenesis analysis. The mechanism by which T368R confers susceptibility, as elucidated by in silico mutagenesis analysis using AmrB-modeled protein structures, is proposed to be due to the location of T368R in a highly conserved region, rather than destabilization of the AmrB protein structure.
Collapse
Affiliation(s)
- Ainulkhir Hussin
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Sheila Nathan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Muhammad Ashraf Shahidan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Mohd Yusof Nor Rahim
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Mohamad Yusof Zainun
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | | | - Nazlina Ibrahim
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia.
| |
Collapse
|
40
|
Filgueiras LA, de Andrade FDCP, Iwao Horita S, Shirsat SD, Achal V, Rai M, Henriques-Pons A, Mendes AN. Analysis of SIKVAV's receptor affinity, pharmacokinetics, and pharmacological characteristics: a matrikine with potent biological function. J Biomol Struct Dyn 2024:1-23. [PMID: 38345036 DOI: 10.1080/07391102.2024.2313709] [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: 09/12/2023] [Accepted: 01/27/2024] [Indexed: 03/08/2025]
Abstract
Matrikines are biologically active peptides generated from fragments fragmentation of extracellular matrix components (ECM) that are functionally distinct from the original full-length molecule. The active matricryptic sites can be unmasked by ECM components enzymatic degradation or multimerization, heterotypic binding, adsorption to other molecules, cell-mediated mechanical forces, exposure to reactive oxygen species, ECM denaturation, and others. Laminin α1-derived peptide (SIKVAV) is a bioactive peptide derived from laminin-111 that participates in tumor development, cell proliferation, angiogenesis in various cell types. SIKVAV has also a potential pharmaceutical activity that may be used for tissue regeneration and bioengineering in Alzheimer's disease and muscular dystrophies. In this work, we made computational analyzes of SIKVAV regarding the ADMET panel, that stands for Administration, Distribution, Metabolism, Excretion, and Toxicity. Docking analyzes using the α3β1 and α6β1 integrin receptors were performed to fill in the gaps in the SIKVAV's signaling pathway and coupling tests showed that SIKVAV can interact with both receptors. Moreover, there is no indication of cytotoxicity, mutagenic or carcinogenic activity, skin or oral sensitivity. Our analysis suggests that SIKVAV has a high probability of interacting with peroxisome proliferator-activated receptor-gamma (NR-PPAR-γ), which has anti-inflammatory activity. The results of bioinformatics can help understand the participation of SIKVAV in homeostasis and influence the understanding of how this peptide can act as a biological asset in the control of dystrophies, neurodegenerative diseases, and tissue engineering.
Collapse
Affiliation(s)
- Livia Alves Filgueiras
- Laboratory of Innovation in Science and Technology - LACITEC, Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | | | - Samuel Iwao Horita
- Laboratory of Innovation in Therapies, Education, and Bioproducts - LITEB, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Shubhangi D Shirsat
- Laboratory of Innovation in Therapies, Education, and Bioproducts - LITEB, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Varenyam Achal
- Environmental Engineering Program, Guangdong Technion - Israel Institute of Technology, Shantou, China
- Technion - Israel Institute of Technology, Haifa, Israel
| | - Mahendra Rai
- Department of Biotechnology, SGB Amravati University, Amravati, India
| | - Andrea Henriques-Pons
- Laboratory of Innovation in Therapies, Education, and Bioproducts - LITEB, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anderson Nogueira Mendes
- Laboratory of Innovation in Science and Technology - LACITEC, Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
41
|
Akermi S, Smaoui S, Chaari M, Elhadef K, Gentile R, Hait M, Roymahapatra G, Mellouli L. Combined in vitro/in silico approaches, molecular dynamics simulations and safety assessment of the multifunctional properties of thymol and carvacrol: A comparative insight. Chem Biodivers 2024; 21:e202301575. [PMID: 38116885 DOI: 10.1002/cbdv.202301575] [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: 10/07/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
Bioactive compounds derived from medicinal plants have acquired immense attentiveness in drug discovery and development. The present study investigated in vitro and predicted in silico the antibacterial, antifungal, and antiviral properties of thymol and carvacrol, and assessed their safety. The performed microbiological assays against Pseudomonas aeruginosa, Escherichia coli, Salmonella enterica Typhimurium revealed that the minimal inhibitory concentration values ranged from (0.078 to 0.312 mg/mL) and the minimal fungicidal concentration against Candida albicans was 0.625 mg/mL. Molecular docking simulations, stipulated that these compounds could inhibit bacterial replication and transcription functions by targeting DNA and RNA polymerases receptors with docking scores varying between (-5.1 to -6.9 kcal/mol). Studied hydroxylated monoterpenes could hinder C. albicans growth by impeding lanosterol 14α-demethylase enzyme and showed a (ΔG=-6.2 and -6.3 kcal/mol). Computational studies revealed that thymol and carvacrol could target the SARS-Cov-2 spike protein of the Omicron variant RBD domain. Molecular dynamics simulations disclosed that these compounds have a stable dynamic behavior over 100 ns as compared to remdesivir. Chemo-computational toxicity prediction using Protox II webserver indicated that thymol and carvacrol could be safely and effectively used as drug candidates to tackle bacterial, fungal, and viral infections as compared to chemical medication.
Collapse
Affiliation(s)
- Sarra Akermi
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Slim Smaoui
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Moufida Chaari
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Khaoula Elhadef
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Rocco Gentile
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Milan Hait
- Department of Chemistry, Dr. C. V. Raman University, Kota, 495113, Bilaspur, India
| | | | - Lotfi Mellouli
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| |
Collapse
|
42
|
Miao C, Cui Y, Li Y, Qi Q, Shang W, Chen H, Gao Y, Yuan R, Long Q, Wu W, Wang X, Yan Z, Jiang Y. Immunoinformatics Prediction and Protective Efficacy of Vaccine Candidate PiuA-PlyD4 Against Streptococcus Pneumoniae. Drug Des Devel Ther 2023; 17:3783-3801. [PMID: 38146490 PMCID: PMC10749580 DOI: 10.2147/dddt.s441302] [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: 09/20/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023] Open
Abstract
Purpose This study was designed to evaluate the immune protective efficacy of the novel Streptococcus pneumoniae (S. pneumoniae) protein vaccine PiuA-PlyD4 through immunoinformatics prediction and in vitro and in vivo experiments. Methods In this study, we conducted immunoinformatics prediction and protection analysis on the fusion protein PiuA-PlyD4. The epitope composition of the vaccine was analyzed based on the prediction of B-cell and helper T-cell epitopes. Meanwhile, the molecular docking of PiuA and TLR2/4 was simulated. After immunizing C57BL/6 mice with the prepared vaccine, the biological safety, immunogenicity and conservation were evaluated. By constructing different infection models and from the aspects of adhesion inhibition and cytokines, the protective effect of the fusion protein vaccine PiuA-PlyD4 on S. pneumoniae infection was explored. Results PiuA-PlyD4 has abundant B-cell and helper T-cell epitopes and shows a high antigenicity score and structural stability. Molecular docking analysis suggested the potential interaction between PiuA and TLR2/4. The specific antibody titer of fusion protein antiserum was as high as (7.81±2.32) ×105. The protective effect of the immunized mice on nasal and lung colonization was significantly better than that of the control group, and the survival rate against S. pneumoniae infection of serotype 3 reached 50%. Cytokine detection showed that the humoral immune response, Th1, Th2 and Th17 cellular immune pathways were all involved in the process. Conclusion The study indicates that PiuA-PlyD4, whether the results are predicted by immunoinformatics or experimentally validated in vivo and in vitro, has good immunogenicity and immunoreactivity and can provide effective protection against S. pneumoniae infection. Therefore, it can be considered a promising prophylactic vaccine candidate for S. pneumoniae.
Collapse
Affiliation(s)
- Chenglin Miao
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Yali Cui
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
- Department of Laboratory Medicine, Meishan Women and Children’s Hospital, Alliance Hospital of West China Second University Hospital, Sichuan University, Meishan, Sichuan Province, People’s Republic of China
- Department of Laboratory Medicine, West China Second University Hospital (Tianfu), Sichuan University/Sichuan Provincial Children’s Hospital, Meishan, Sichuan Province, People’s Republic of China
| | - Yingying Li
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Qianqian Qi
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Wenling Shang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Huilian Chen
- Department of Laboratory Medicine, Meishan Women and Children’s Hospital, Alliance Hospital of West China Second University Hospital, Sichuan University, Meishan, Sichuan Province, People’s Republic of China
- Department of Laboratory Medicine, West China Second University Hospital (Tianfu), Sichuan University/Sichuan Provincial Children’s Hospital, Meishan, Sichuan Province, People’s Republic of China
| | - Yujie Gao
- Department of Laboratory Medicine, Meishan Women and Children’s Hospital, Alliance Hospital of West China Second University Hospital, Sichuan University, Meishan, Sichuan Province, People’s Republic of China
- Department of Laboratory Medicine, West China Second University Hospital (Tianfu), Sichuan University/Sichuan Provincial Children’s Hospital, Meishan, Sichuan Province, People’s Republic of China
| | - Ruomei Yuan
- Department of Laboratory Medicine, Meishan Women and Children’s Hospital, Alliance Hospital of West China Second University Hospital, Sichuan University, Meishan, Sichuan Province, People’s Republic of China
- Department of Laboratory Medicine, West China Second University Hospital (Tianfu), Sichuan University/Sichuan Provincial Children’s Hospital, Meishan, Sichuan Province, People’s Republic of China
| | - Qichen Long
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Wenjing Wu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Xia Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Ziyi Yan
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
43
|
Binbay FA, Rathod DC, George AAP, Imhof D. Quality Assessment of Selected Protein Structures Derived from Homology Modeling and AlphaFold. Pharmaceuticals (Basel) 2023; 16:1662. [PMID: 38139789 PMCID: PMC10747200 DOI: 10.3390/ph16121662] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
With technology advancing, many prediction algorithms have been developed to facilitate the modeling of inherently dynamic and flexible macromolecules such as proteins. Improvements in the prediction of protein structures have attracted a great deal of attention due to the advantages they offer, e.g., in drug design. While trusted experimental methods, such as X-ray crystallography, NMR spectroscopy, and electron microscopy, are preferred structure analysis techniques, in silico approaches are also being widely used. Two computational methods, which are on opposite ends of the spectrum with respect to their modus operandi, i.e., homology modeling and AlphaFold, have been established to provide high-quality structures. Here, a comparative study of the quality of structures either predicted by homology modeling or by AlphaFold is presented based on the characteristics determined by experimental studies using structure validation servers to fulfill the purpose. Although AlphaFold is able to predict high-quality structures, high-confidence parts are sometimes observed to be in disagreement with experimental data. On the other hand, while the structures obtained from homology modeling are successful in incorporating all aspects of the experimental structure used as a template, this method may struggle to accurately model a structure in the absence of a suitable template. In general, although both methods produce high-quality models, the criteria by which they are superior to each other are different and thus discussed in detail.
Collapse
Affiliation(s)
- Furkan Ayberk Binbay
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Dhruv Chetanbhai Rathod
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | | | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
44
|
Behairy MY, Eid RA, Otifi HM, Mohammed HM, Alshehri MA, Asiri A, Aldehri M, Zaki MSA, Darwish KM, Elhady SS, El-Shaer NH, Eldeen MA. Unraveling Extremely Damaging IRAK4 Variants and Their Potential Implications for IRAK4 Inhibitor Efficacy. J Pers Med 2023; 13:1648. [PMID: 38138875 PMCID: PMC10744719 DOI: 10.3390/jpm13121648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/24/2023] Open
Abstract
Interleukin-1-receptor-associated kinase 4 (IRAK4) possesses a crucial function in the toll-like receptor (TLR) signaling pathway, and the dysfunction of this molecule could lead to various infectious and immune-related diseases in addition to cancers. IRAK4 genetic variants have been linked to various types of diseases. Therefore, we conducted a comprehensive analysis to recognize the missense variants with the most damaging impacts on IRAK4 with the employment of diverse bioinformatics tools to study single-nucleotide polymorphisms' effects on function, stability, secondary structures, and 3D structure. The residues' location on the protein domain and their conservation status were investigated as well. Moreover, docking tools along with structural biology were engaged in analyzing the SNPs' effects on one of the developed IRAK4 inhibitors. By analyzing IRAK4 gene SNPs, the analysis distinguished ten variants as the most detrimental missense variants. All variants were situated in highly conserved positions on an important protein domain. L318S and L318F mutations were linked to changes in IRAK4 secondary structures. Eight SNPs were revealed to have a decreasing effect on the stability of IRAK4 via both I-Mutant 2.0 and Mu-Pro tools, while Mu-Pro tool identified a decreasing effect for the G198E SNP. In addition, detrimental effects on the 3D structure of IRAK4 were also discovered for the selected variants. Molecular modeling studies highlighted the detrimental impact of these identified SNP mutant residues on the druggability of the IRAK4 ATP-binding site towards the known target inhibitor, HG-12-6, as compared to the native protein. The loss of important ligand residue-wise contacts, altered protein global flexibility, increased steric clashes, and even electronic penalties at the ligand-binding site interfaces were all suggested to be associated with SNP models for hampering the HG-12-6 affinity towards IRAK4 target protein. This given model lays the foundation for the better prediction of various disorders relevant to IRAK4 malfunction and sheds light on the impact of deleterious IRAK4 variants on IRAK4 inhibitor efficacy.
Collapse
Affiliation(s)
- Mohammed Y. Behairy
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt;
| | - Refaat A. Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha P.O. Box 61421, Saudi Arabia; (R.A.E.); (H.M.O.)
| | - Hassan M. Otifi
- Department of Pathology, College of Medicine, King Khalid University, Abha P.O. Box 61421, Saudi Arabia; (R.A.E.); (H.M.O.)
| | - Heitham M. Mohammed
- Department of Anatomy, College of Medicine, King Khalid University, Abha P.O. Box 61421, Saudi Arabia; (H.M.M.); (M.A.); (M.S.A.Z.)
| | - Mohammed A. Alshehri
- Department of Child Health, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia; (M.A.A.)
| | - Ashwag Asiri
- Department of Child Health, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia; (M.A.A.)
| | - Majed Aldehri
- Department of Anatomy, College of Medicine, King Khalid University, Abha P.O. Box 61421, Saudi Arabia; (H.M.M.); (M.A.); (M.S.A.Z.)
| | - Mohamed Samir A. Zaki
- Department of Anatomy, College of Medicine, King Khalid University, Abha P.O. Box 61421, Saudi Arabia; (H.M.M.); (M.A.); (M.S.A.Z.)
| | - Khaled M. Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Nahla H. El-Shaer
- Department of Zoology, Faculty of Science, Zagazig University, Zagazig 44511, Egypt;
| | - Muhammad Alaa Eldeen
- Department of Zoology, Faculty of Science, Zagazig University, Zagazig 44511, Egypt;
| |
Collapse
|
45
|
Bærentsen RL, Nielsen SV, Skjerning RB, Lyngsø J, Bisiak F, Pedersen JS, Gerdes K, Sørensen MA, Brodersen DE. Structural basis for kinase inhibition in the tripartite E. coli HipBST toxin-antitoxin system. eLife 2023; 12:RP90400. [PMID: 37929938 PMCID: PMC10627512 DOI: 10.7554/elife.90400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
Many bacteria encode multiple toxin-antitoxin (TA) systems targeting separate, but closely related, cellular functions. The toxin of the Escherichia coli hipBA system, HipA, is a kinase that inhibits translation via phosphorylation of glutamyl-tRNA synthetase. Enteropathogenic E. coli O127:H6 encodes the hipBA-like, tripartite TA system; hipBST, in which the HipT toxin specifically targets the tryptophanyl-tRNA synthetase, TrpS. Notably, in the tripartite system, the function as antitoxin has been taken over by the third protein, HipS, but the molecular details of how activity of HipT is inhibited remain poorly understood. Here, we show that HipBST is structurally different from E. coli HipBA and that the unique HipS protein, which is homologous to the N-terminal subdomain of HipA, inhibits the kinase through insertion of a conserved Trp residue into the active site. We also show how auto-phosphorylation at two conserved sites in the kinase toxin serve different roles and affect the ability of HipS to neutralize HipT. Finally, solution structural studies show how phosphorylation affects overall TA complex flexibility.
Collapse
Affiliation(s)
- René L Bærentsen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Stine V Nielsen
- Department of Biology, University of CopenhagenCopenhagenDenmark
| | | | - Jeppe Lyngsø
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO)AarhusDenmark
| | - Francesco Bisiak
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Jan Skov Pedersen
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO)AarhusDenmark
| | | | | | - Ditlev E Brodersen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| |
Collapse
|
46
|
Bappi MH, Prottay AAS, Al-Khafaji K, Akbor MS, Hossain MK, Islam MS, Asha AI, Medeiros CR, Tahim CM, Lucetti ECP, Coutinho HDM, Kamli H, Islam MT. Antiemetic effects of sclareol, possibly through 5-HT 3 and D 2 receptor interaction pathways: In-vivo and in-silico studies. Food Chem Toxicol 2023; 181:114068. [PMID: 37863383 DOI: 10.1016/j.fct.2023.114068] [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: 06/05/2023] [Revised: 09/12/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Emesis is a complex physiological phenomenon that serves as a defense against numerous toxins, stressful situations, adverse medication responses, chemotherapy, and movement. Nevertheless, preventing emesis during chemotherapy or other situations is a significant issue for researchers. Hence, the majority view contends that successfully combining therapy is the best course of action. In-vivo analysis offers a more comprehensive grasp of how compounds behave within a complex biological environment, whereas in-silico evaluation refers to the use of computational models to forecast biological interactions. OBJECTIVES The objectives of the present study were to evaluate the effects of Sclareol (SCL) on copper sulphate-induced emetic chicks and to investigate the combined effects of these compounds using a conventional co-treatment approach and in-silico study. METHODS SCL (5, 10, and 15 mg/kg) administered orally with or without pre-treatment with anti-emetic drugs (Ondansetron (ODN): 24 mg/kg, Domperidone (DOM): 80 mg/kg, Hyoscine butylbromide (HYS): 100 mg/kg, and Promethazine hydrochloride (PRO): 100 mg/kg) to illustrate the effects and the potential involvement with 5HT3, D2, M3/AChM, H1, or NK1 receptors by SCL. Furthermore, an in-silico analysis was conducted to forecast the role of these receptors in the emetic process. RESULTS The results suggest that SCL exerted a dose-dependent anti-emetic effect on the chicks. Pretreatment with SCL-10 significantly minimized the number of retches and lengthened the emesis tendency of the experimental animals. SCL-10 significantly increased the anti-emetic effects of ODN and DOM. However, compared to the ODN-treated group, (SCL-10 + ODN) group considerably (p < 0.0001) extended the latency duration (109.40 ± 1.03 s) and significantly (p < 0.01) decreased the number of retches (20.00 ± 0.70), indicating an anti-emetic effect on the test animals. In in-silico analysis, SCL exhibited promising binding affinities with suggesting receptors. CONCLUSION SCL-10 exerted an inhibitory-like effect on emetic chicks, probably through the interaction of the 5HT3 and D2 receptors. Further studies are highly appreciated to validate this study and determine the precise mechanism(s) behind the anti-emetic effects of SCL. We expect that SCL-10 may be utilized as an antiemetic treatment in a single dosage form or that it may function as a synergist with other traditional medicines.
Collapse
Affiliation(s)
- Mehedi Hasan Bappi
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Abdullah Al Shamsh Prottay
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Khattab Al-Khafaji
- Department of Environmental Science, College of Energy and Environmental Science, Al-Karkh University of Science, Baghdad, 10081, Iraq
| | - Md Showkoth Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Muhammad Kamal Hossain
- School of Pharmacy, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Department of Pharmacy, University of Science & Technology Chittagong, Chittagong, 4202, Bangladesh
| | - Md Shahazul Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Afia Ibnath Asha
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Cassio Rocha Medeiros
- CECAPE College, Av. Padre Cícero, 3917 - São José, Juazeiro Do Norte, CE, 63024-015, Brazil
| | - Catarina Martins Tahim
- CECAPE College, Av. Padre Cícero, 3917 - São José, Juazeiro Do Norte, CE, 63024-015, Brazil
| | | | - Henrique Douglas Melo Coutinho
- Department of Biological Chemistry, Laboratory of Microbiology and Molecular Biology, Regional University of Cariri, Crato, CE, 63105-000, Brazil.
| | - Hossam Kamli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
47
|
Rosenberg AA, Yehishalom N, Marx A, Bronstein AM. An amino-domino model described by a cross-peptide-bond Ramachandran plot defines amino acid pairs as local structural units. Proc Natl Acad Sci U S A 2023; 120:e2301064120. [PMID: 37878722 PMCID: PMC10623034 DOI: 10.1073/pnas.2301064120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 08/24/2023] [Indexed: 10/27/2023] Open
Abstract
Protein structure, both at the global and local level, dictates function. Proteins fold from chains of amino acids, forming secondary structures, α-helices and β-strands, that, at least for globular proteins, subsequently fold into a three-dimensional structure. Here, we show that a Ramachandran-type plot focusing on the two dihedral angles separated by the peptide bond, and entirely contained within an amino acid pair, defines a local structural unit. We further demonstrate the usefulness of this cross-peptide-bond Ramachandran plot by showing that it captures β-turn conformations in coil regions, that traditional Ramachandran plot outliers fall into occupied regions of our plot, and that thermophilic proteins prefer specific amino acid pair conformations. Further, we demonstrate experimentally that the effect of a point mutation on backbone conformation and protein stability depends on the amino acid pair context, i.e., the identity of the adjacent amino acid, in a manner predictable by our method.
Collapse
Affiliation(s)
- Aviv A. Rosenberg
- Department of Computer Science, Technion–Israel Institute of Technology, Haifa32000, Israel
| | - Nitsan Yehishalom
- Faculty of Biology, Technion–Israel Institute of Technology, Haifa32000, Israel
| | - Ailie Marx
- Department of Computer Science, Technion–Israel Institute of Technology, Haifa32000, Israel
| | - Alex M. Bronstein
- Department of Computer Science, Technion–Israel Institute of Technology, Haifa32000, Israel
| |
Collapse
|
48
|
Abdul Kadir FFN, Che Nordin MA, S M N Mydin RB, Choong YS, Che Omar MT. Molecular interaction analysis of anti-IL-8 scFv-10F8-6His against IL-8 monomer through molecular docking and molecular dynamic simulations. J Biomol Struct Dyn 2023; 42:12293-12303. [PMID: 37837430 DOI: 10.1080/07391102.2023.2269254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023]
Abstract
Elevated interleukin 8 (IL-8) expression has been linked to unfavorable outcomes in a range of inflammatory conditions, such as rheumatoid arthritis, psoriasis, and cancer. The human monoclonal antibody (HuMab) 10F8 and the hybridoma 35B11-B bind to an epitope on human IL-8, respectively. 10F8 inhibited interaction between IL-8 and neutrophils in eczema and pustulosis palmoplantaris patients while 35B11-B decreased size lesion in rat model. The binding interaction of monoclonal antibodies and IL-8, especially how complementarity-determining region (CDR) loops could bind the N-terminal of IL-8, has not been fully deliberated at molecular-level. Here, we used a combination of molecular docking, heated and long coarse-grained molecular dynamics simulations to identify key residues of established interaction. Based on heated MD simulation, docked pose of complexes generated by ClusPro showed good binding stability throughout of 70 ns simulation. Based on long molecular dynamic simulations, key residues for the binding were identified throughout of 1000 ns simulation. TYR-53, ASP-99, and ARG-100 of heavy chain CDR together with TYR-33 of light chain CDR are among the highest contributing energy residues within the binding interaction. Meanwhile, LYS11 and TYR13 of IL-8 are important for the determination of overall binding energy. Furthermore, the result of decomposition residues analysis is in good agreement with the interaction analysis data. Current study provides a list of important interacting residues and further scrutiny on these residues is essential for future development and design of a new and stable recombinant antibody against IL-8.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Muhamad Alif Che Nordin
- Biological Section, School of Distance Education, Universiti Sains Malaysia, Penang, Malaysia
| | - Rabiatul Basria S M N Mydin
- Biomedical Sciences Department, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang, Malaysia
| | | |
Collapse
|
49
|
Khattak AA, Qian J, Xu L, Tomah AA, Ibrahim E, Khan MZI, Ahmed T, Hatamleh AA, Al-Dosary MA, Ali HM, Li B. Precision drug design against Acidovorax oryzae: leveraging bioinformatics to combat rice brown stripe disease. Front Cell Infect Microbiol 2023; 13:1225285. [PMID: 37886665 PMCID: PMC10598866 DOI: 10.3389/fcimb.2023.1225285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
Bacterial brown stripe disease caused by Acidovorax oryzae is a major threat to crop yields, and the current reliance on pesticides for control is unsustainable due to environmental pollution and resistance. To address this, bacterial-based ligands have been explored as a potential treatment solution. In this study, we developed a protein-protein interaction (PPI) network for A. oryzae by utilizing shared differentially expressed genes (DEGs) and the STRING database. Using a maximal clique centrality (MCC) approach through CytoHubba and Network Analyzer, we identified hub genes within the PPI network. We then analyzed the genomic data of the top 10 proteins, and further narrowed them down to 2 proteins by utilizing betweenness, closeness, degree, and eigenvector studies. Finally, we used molecular docking to screen 100 compounds against the final two proteins (guaA and metG), and Enfumafungin was selected as a potential treatment for bacterial resistance caused by A. oryzae based on their binding affinity and interaction energy. Our approach demonstrates the potential of utilizing bioinformatics and molecular docking to identify novel drug candidates for precision treatment of bacterial brown stripe disease caused by A. oryzae, paving the way for more targeted and sustainable control strategies. The efficacy of Enfumafungin in inhibiting the growth of A. oryzae strain RS-1 was investigated through both computational and wet lab methods. The models of the protein were built using the Swiss model, and their accuracy was confirmed via a Ramachandran plot. Additionally, Enfumafungin demonstrated potent inhibitory action against the bacterial strain, with an MIC of 100 µg/mL, reducing OD600 values by up to 91%. The effectiveness of Enfumafungin was further evidenced through agar well diffusion assays, which exhibited the highest zone of inhibition at 1.42 cm when the concentration of Enfumafungin was at 100 µg/mL. Moreover, Enfumafungin was also able to effectively reduce the biofilm of A. oryzae RS-1 in a concentration-dependent manner. The swarming motility of A. oryzae RS-1 was also found to be significantly inhibited by Enfumafungin. Further validation through TEM observation revealed that bacterial cells exposed to Enfumafungin displayed mostly red fluorescence, indicating destruction of the bacterial cell membrane.
Collapse
Affiliation(s)
- Arif Ali Khattak
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Jiahui Qian
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Lihui Xu
- Institute of Eco-Environmental Protection, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Ali Athafah Tomah
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Plant Protection, College of Agriculture, University of Misan, AL-Amarah, Iraq
| | - Ezzeldin Ibrahim
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Department of Vegetable Diseases Research, Plant Pathology Research Institute, Agriculture Research Centre, Giza, Egypt
| | | | - Temoor Ahmed
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Xianghu Laboratory, Hangzhou, China
| | - Ashraf Atef Hatamleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Hayssam M. Ali
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Bin Li
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| |
Collapse
|
50
|
van Alen I, Chikunova A, van Zanten DB, de Block AA, Timmer M, Brünle S, Ubbink M. Asp179 in the class A β-lactamase from Mycobacterium tuberculosis is a conserved yet not essential residue due to epistasis. FEBS J 2023; 290:4933-4949. [PMID: 37335937 DOI: 10.1111/febs.16892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/22/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023]
Abstract
Conserved residues are often considered essential for function, and substitutions in such residues are expected to have a negative influence on the properties of a protein. However, mutations in a few highly conserved residues of the β-lactamase from Mycobacterium tuberculosis, BlaC, were shown to have no or only limited negative effect on the enzyme. One such mutant, D179N, even conveyed increased ceftazidime resistance upon bacterial cells, while displaying good activity against penicillins. The crystal structures of BlaC D179N in resting state and in complex with sulbactam reveal subtle structural changes in the Ω-loop as compared to the structure of wild-type BlaC. Introducing this mutation in four other β-lactamases, CTX-M-14, KPC-2, NMC-A and TEM-1, resulted in decreased antibiotic resistance for penicillins and meropenem. The results demonstrate that the Asp in position 179 is generally essential for class A β-lactamases but not for BlaC, which can be explained by the importance of the interaction with the side chain of Arg164 that is absent in BlaC. It is concluded that Asp179 though conserved is not essential in BlaC, as a consequence of epistasis.
Collapse
Affiliation(s)
- Ilona van Alen
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | | - Danny B van Zanten
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Amber A de Block
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Monika Timmer
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Steffen Brünle
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Marcellus Ubbink
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| |
Collapse
|