151
|
Ojima-Kato T. Advances in recombinant protein production in microorganisms and functional peptide tags. Biosci Biotechnol Biochem 2024; 89:1-10. [PMID: 39479788 DOI: 10.1093/bbb/zbae147] [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: 09/08/2024] [Accepted: 10/13/2024] [Indexed: 12/24/2024]
Abstract
Recombinant protein production in prokaryotic and eukaryotic cells is a fundamental technology for both research and industry. Achieving efficient protein synthesis is key to accelerating the discovery, characterization, and practical application of proteins. This review focuses on recent advances in recombinant protein production and strategies for more efficient protein production, especially using Escherichia coli and Saccharomyces cerevisiae. Additionally, this review summarizes the development of various functional peptide tags that can be employed for protein production, modification, and purification, including translation-enhancing peptide tags developed by our research group.
Collapse
Affiliation(s)
- Teruyo Ojima-Kato
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
152
|
Mathur A, Ghosh R, Nunes-Alves A. Recent Progress in Modeling and Simulation of Biomolecular Crowding and Condensation Inside Cells. J Chem Inf Model 2024; 64:9063-9081. [PMID: 39660892 DOI: 10.1021/acs.jcim.4c01520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Macromolecular crowding in the cellular cytoplasm can potentially impact diffusion rates of proteins, their intrinsic structural stability, binding of proteins to their corresponding partners as well as biomolecular organization and phase separation. While such intracellular crowding can have a large impact on biomolecular structure and function, the molecular mechanisms and driving forces that determine the effect of crowding on dynamics and conformations of macromolecules are so far not well understood. At a molecular level, computational methods can provide a unique lens to investigate the effect of macromolecular crowding on biomolecular behavior, providing us with a resolution that is challenging to reach with experimental techniques alone. In this review, we focus on the various physics-based and data-driven computational methods developed in the past few years to investigate macromolecular crowding and intracellular protein condensation. We review recent progress in modeling and simulation of biomolecular systems of varying sizes, ranging from single protein molecules to the entire cellular cytoplasm. We further discuss the effects of macromolecular crowding on different phenomena, such as diffusion, protein-ligand binding, and mechanical and viscoelastic properties, such as surface tension of condensates. Finally, we discuss some of the outstanding challenges that we anticipate the community addressing in the next few years in order to investigate biological phenomena in model cellular environments by reproducing in vivo conditions as accurately as possible.
Collapse
Affiliation(s)
- Apoorva Mathur
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Rikhia Ghosh
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877, United States
| | - Ariane Nunes-Alves
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
| |
Collapse
|
153
|
Cresca S, Parise A, Magistrato A. Assessing the Mechanism of Rac1b: An All-Atom Simulation Study of the Alternative Spliced Variant of Rac1 Small Rho GTPase. J Chem Inf Model 2024; 64:9474-9486. [PMID: 39632743 DOI: 10.1021/acs.jcim.4c01376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The Rho GTPase family plays a key role in cell migration, cytoskeletal dynamics, and intracellular signaling. Rac1 and its splice variant Rac1b, characterized by the insertion of an Extraloop, are frequently associated with cancer. These small GTPases switch between an active GTP-bound state and an inactive GDP-bound state, a process that is regulated by specific protein modulators. Among them, the Guanine nucleotide exchange factor (GEF) protein DOCK5 specifically targets Rho GTPases, promoting their activation by facilitating the exchange of GDP for GTP. In this study, we performed cumulative 10-μs-long all-atom molecular dynamics simulations of Rac1 and Rac1b, in isolation and in complex with DOCK5 and ELMO1, to investigate the impact of the Rac1b Extraloop. Our findings reveal that this Extraloop decreases the GDP residence time as compared to Rac1, mimicking the effect of accelerated GDP/GTP exchange induced by DOCK5. Furthermore, both Rac1b Extraloop and the ELMO1 protein stabilize the GTPase/DOCK5 complex, contributing to facilitate GDP dissociation. This shifts the balance between the GPT- and GDP-bound state of Rac1b toward the active GTP-bound state, sending a prooncogenic signal. Besides broadening our understanding of the biological functions of small Rho GTPases, this study provides key information to exploit a previously unexplored therapeutic niche to counter Rac1b-associated cancer.
Collapse
Affiliation(s)
- Sofia Cresca
- Theory Department, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Consiglio Nazionale delle Ricerche (CNR)-IOM, c/o International School for Advanced Studies (SISSA/ISAS), via Bonomea 265, 34136 Trieste, Italy
| | - Angela Parise
- Consiglio Nazionale delle Ricerche (CNR)-IOM, c/o International School for Advanced Studies (SISSA/ISAS), via Bonomea 265, 34136 Trieste, Italy
| | - Alessandra Magistrato
- Consiglio Nazionale delle Ricerche (CNR)-IOM, c/o International School for Advanced Studies (SISSA/ISAS), via Bonomea 265, 34136 Trieste, Italy
| |
Collapse
|
154
|
Raza Z, Arockiaraj M, Maaran A, Shalini AJ. A comparative study of topological entropy characterization and graph energy prediction for Marta variants of covalent organic frameworks. Front Chem 2024; 12:1511678. [PMID: 39758156 PMCID: PMC11696151 DOI: 10.3389/fchem.2024.1511678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025] Open
Abstract
Covalent organic frameworks are a novel class of porous polymers, notable for their crystalline structure, intricate frameworks, defined pore sizes, and capacity for structural design, synthetic control, and functional customization. This paper provides a comprehensive analysis of graph entropies and hybrid topological descriptors, derived from geometric, harmonic, and Zagreb indices. These descriptors are applied to study two variations of Marta covalent organic frameworks based on contorted hexabenzocoronenes. We also conduct a comparative analysis using scaled entropies, offering refined tools for assessing the intrinsic topologies of these networks. Additionally, these hybrid descriptors are used to develop statistical models for predicting graph energy in higher-dimensional Marta-COFs.
Collapse
Affiliation(s)
- Zahid Raza
- Department of Mathematics, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Aravindan Maaran
- Department of Mathematics, Loyola College, University of Madras, Chennai, India
| | - Arul Jeya Shalini
- Department of Mathematics, Women’s Christian College, Chennai, India
| |
Collapse
|
155
|
Grzyb T, Szulc J. Deciphering Molecular Mechanisms and Diversity of Plant Holobiont Bacteria: Microhabitats, Community Ecology, and Nutrient Acquisition. Int J Mol Sci 2024; 25:13601. [PMID: 39769364 PMCID: PMC11677812 DOI: 10.3390/ijms252413601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
While gaining increasing attention, plant-microbiome-environment interactions remain insufficiently understood, with many aspects still underexplored. This article explores bacterial biodiversity across plant compartments, including underexplored niches such as seeds and flowers. Furthermore, this study provides a systematic dataset on the taxonomic structure of the anthosphere microbiome, one of the most underexplored plant niches. This review examines ecological processes driving microbial community assembly and interactions, along with the discussion on mechanisms and diversity aspects of processes concerning the acquisition of nitrogen, phosphorus, potassium, and iron-elements essential in both molecular and ecological contexts. These insights are crucial for advancing molecular biology, microbial ecology, environmental studies, biogeochemistry, and applied studies. Moreover, the authors present the compilation of molecular markers for discussed processes, which will find application in (phylo)genetics, various (meta)omic approaches, strain screening, and monitoring. Such a review can be a valuable source of information for specialists in the fields concerned and for applied researchers, contributing to developments in sustainable agriculture, environmental protection, and conservation biology.
Collapse
Affiliation(s)
| | - Justyna Szulc
- Department of Environmental Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Wolczanska 171/173, 90-924 Lodz, Poland;
| |
Collapse
|
156
|
Del Giudice A, Del Giudice D, Spatola E, Alemanno V, Galantini L, Di Stefano S. An albumin unfolding and refolding cycle induced by a time-controlled pH jump. Org Biomol Chem 2024; 23:118-125. [PMID: 39359130 DOI: 10.1039/d4ob01289e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Given the intimate connection between the structure and function of biological macromolecules, the ability to temporally control their unfolding-refolding process enables temporal regulation over specific functionalities, potentially applicable in innovative domains, including the construction of protein-based actuators or programmable catalysis and drug release in complex biotechnological processes. We show here how a temporally controlled protein unfolding-refolding cycle can be coupled in time with programmed pH sequences achieved through the spontaneous decomposition of an activated carboxylic acid. Specifically, we illustrate this process at the molecular level using albumin, the most prevalent protein found in plasma, for which a temporary shift from native to unfolded forms is promoted using nitroacetic acid, able to undergo base-catalysed decarboxylation when solubilized in water solution. As detected by small angle X-ray scattering and intrinsic tryptophan fluorescence, starting from the protein in its native form, the acid addition triggers unfolding to a partially denatured state and a subsequent time-tunable pH rise with gradual refolding that recapitulates the intermediate steps detected at the same pH values by static acidification, fitting within a framework of full reversibility of the structural changes as a function of the protein protonation state. At the end of the pH jump, the native structure is fully recovered, making this method a chemical tool to achieve a complete protein conformational sequence programmed in the timeframe of minutes without further intervention.
Collapse
Affiliation(s)
- Alessandra Del Giudice
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Daniele Del Giudice
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Emanuele Spatola
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Valentina Alemanno
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Luciano Galantini
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Stefano Di Stefano
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
157
|
Zhou Q, Lok SM. Visualizing the virus world inside the cell by cryo-electron tomography. J Virol 2024; 98:e0108523. [PMID: 39494908 PMCID: PMC11650999 DOI: 10.1128/jvi.01085-23] [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] [Indexed: 11/05/2024] Open
Abstract
Structural studies on purified virus have revealed intricate architectures, but there is little structural information on how viruses interact with host cells in situ. Cryo-focused ion beam (FIB) milling and cryo-electron tomography (cryo-ET) have emerged as revolutionary tools in structural biology to visualize the dynamic conformational of viral particles and their interactions with host factors within infected cells. Here, we review the state-of-the-art cryo-ET technique for in situ viral structure studies and highlight exemplary studies that showcase the remarkable capabilities of cryo-ET in capturing the dynamic virus-host interaction, advancing our understanding of viral infection and pathogenesis.
Collapse
Affiliation(s)
- Qunfei Zhou
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Shee-Mei Lok
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Biological Sciences, Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
158
|
Phillips M, Cook ED, Marunde MR, Tonelli M, Khan L, Henrickson A, Lignos JM, Stein JL, Stein GS, Frietze S, Demeler B, Glass KC. The CECR2 bromodomain displays distinct binding modes to select for acetylated histone proteins versus non-histone ligands. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.09.627393. [PMID: 39713312 PMCID: PMC11661176 DOI: 10.1101/2024.12.09.627393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The cat eye syndrome chromosome region candidate 2 (CECR2) protein is an epigenetic regulator involved in chromatin remodeling and transcriptional control. The CECR2 bromodomain (CECR2-BRD) plays a pivotal role in directing the activity of CECR2 through its capacity to recognize and bind acetylated lysine residues on histone proteins. This study elucidates the binding specificity and structural mechanisms of CECR2-BRD interactions with both histone and non-histone ligands, employing techniques such as isothermal titration calorimetry (ITC), nuclear magnetic resonance (NMR) spectroscopy, and a high-throughput peptide assay. The CECR2-BRD selectively binds acetylated histone H3 and H4 ligands, exhibiting a preference for multi-acetylated over mono-acetylated targets. The highest affinity was observed for tetra-acetylated histone H4. Neighboring post-translational modifications, including methylation and phosphorylation, modulate acetyllysine recognition, with significant effects observed for histone H3 ligands. Additionally, this study explored the interaction of the CECR2-BRD with the acetylated RelA subunit of NF-κB, a pivotal transcription factor in inflammatory signaling. Dysregulated NF-κB signaling is implicated in numerous pathologies, including cancer progression, with acetylation of RelA at lysine 310 (K310ac) being critical for its transcriptional activity. Recent evidence linking the CECR2-BRD to RelA suggests it plays a role in inflammatory and metastatic pathways, underscoring the need to understand the molecular basis of this interaction. We found the CECR2-BRD binds to acetylated RelA with micromolar affinity, and uses a distinctive binding mode to recognize this non-histone ligand. These results provide new insight on the role of CECR2 in regulating NF-κB-mediated inflammatory pathways. Functional mutagenesis of critical residues, such as Asn514 and Asp464, highlight their roles in ligand specificity and binding dynamics. Notably, the CECR2-BRD remained monomeric in solution and exhibited differential conformational responses upon ligand binding, suggesting adaptive recognition mechanisms. Furthermore, the CECR2-BRD exclusively interacts with nucleosome substrates containing multi-acetylated histones, emphasizing its role in transcriptional activation within euchromatic regions. These findings position the CECR2-BRD as a key chromatin reader and a promising therapeutic target for modulating transcriptional and inflammatory processes, particularly through the development of selective bromodomain inhibitors.
Collapse
Affiliation(s)
- Margaret Phillips
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Elizabeth D. Cook
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | | | - Marco Tonelli
- National Magnetic Resonance Facility at Madison and Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Laiba Khan
- EpiCypher Inc., Durham, North Carolina 27709, USA
| | - Amy Henrickson
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - James M. Lignos
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Janet L. Stein
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
- University of Vermont Cancer Center, Burlington, VT 05405, USA
| | - Gary S. Stein
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
- University of Vermont Cancer Center, Burlington, VT 05405, USA
| | - Seth Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
- University of Vermont Cancer Center, Burlington, VT 05405, USA
| | - Borries Demeler
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Karen C. Glass
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
- University of Vermont Cancer Center, Burlington, VT 05405, USA
| |
Collapse
|
159
|
Singh A, Sharma A, Singh K, Kaur K, Mohana P, Prajapati J, Kaur U, Goswami D, Arora S, Chadha R, Singh Bedi PM. Development of coumarin-inspired bifunctional hybrids as a new class of anti-Alzheimer's agents with potent in vivo efficacy. RSC Med Chem 2024:d4md00782d. [PMID: 39790122 PMCID: PMC11707525 DOI: 10.1039/d4md00782d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
Considering the multifactorial and complex nature of Alzheimer's disease and the requirement of an optimum multifunctional anti-Alzheimer's agent, a series of triazole tethered coumarin-eugenol hybrid molecules was designed as potential multifunctional anti-Alzheimer's agents using donepezil and a template. The designed hybrid molecules were synthesized via a click chemistry approach and preliminarily screened for cholinesterase and Aβ1-42 aggregation inhibition. Among them, AS15 emerged as a selective inhibitor of AChE (IC50 = 0.047 μM) over butyrylcholinesterase (BuChE: IC50 ≥ 10 μM) with desired Aβ1-42 aggregation inhibition (72.21% at 50 μM) properties. In addition, AS15 showed protective effects against DNA damage caused by hydroxyl radicals originating from H2O2. Molecular docking and simulation studies confirmed the favorable interactions of AChE and the Aβ1-42 monomer desired for their inhibition. AS15 exhibited an LD50 value of 300 mg kg-1 and showed significant improvements in memory and learning behavior in scopolamine-induced cognition impairment mouse-based animal models (Y-maze test and Morris water maze test) for behavioral analysis. Overall outcomes suggest AS15 as a potential preclinical multifunctional candidate for the management of Alzheimer's disease, and it serves as a promising lead for further development of potent and safer multifunctional anti-Alzheimer's agents.
Collapse
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Aman Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Pallvi Mohana
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Jignesh Prajapati
- Department of Biochemistry & Forensic Science, University School of Sciences, Gujrat University Ahmedabad Gujrat 380009 India
| | - Uttam Kaur
- University School of Business Management, Chandigarh University Gharuan 140413 India
| | - Dweipayan Goswami
- Department of Microbiology & Biotechnology, University School of Sciences, Gujrat University Ahmedabad Gujrat 380009 India
| | - Saroj Arora
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Renu Chadha
- University Institute of Pharmaceutical Sciences, Punjab University Chandigarh 160014 India
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
- Drug and Pollution testing Laboratory, Guru Nanak Dev University Amritsar Punjab 143005 India
| |
Collapse
|
160
|
Choi AA, Zhou CY, Tabo A, Heald R, Xu K. Single-molecule diffusivity quantification in Xenopus egg extracts elucidates physicochemical properties of the cytoplasm. Proc Natl Acad Sci U S A 2024; 121:e2411402121. [PMID: 39636857 DOI: 10.1073/pnas.2411402121] [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: 06/08/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024] Open
Abstract
The living cell creates a unique internal molecular environment that is challenging to characterize. By combining single-molecule displacement/diffusivity mapping (SMdM) with physiologically active extracts prepared from Xenopus laevis eggs, we sought to elucidate molecular properties of the cytoplasm. Quantification of the diffusion coefficients of 15 diverse proteins in extract showed that, compared to in water, negatively charged proteins diffused ~50% slower, while diffusion of positively charged proteins was reduced by ~80 to 90%. Adding increasing concentrations of salt progressively alleviated the suppressed diffusion observed for positively charged proteins, signifying electrostatic interactions within a predominately negatively charged macromolecular environment. To investigate the contribution of RNA, an abundant, negatively charged component of cytoplasm, extracts were treated with ribonuclease, which resulted in low diffusivity domains indicative of aggregation, likely due to the liberation of positively charged RNA-binding proteins such as ribosomal proteins, since this effect could be mimicked by adding positively charged polypeptides. Interestingly, in extracts prepared under typical conditions that inhibit actin polymerization, negatively charged proteins of different sizes showed similar diffusivity suppression consistent with our separately measured 2.22-fold higher viscosity of extract over water. Restoring or enhancing actin polymerization progressively suppressed the diffusion of larger proteins, recapitulating behaviors observed in cells. Together, these results indicate that molecular interactions in the crowded cell are defined by an overwhelmingly negatively charged macromolecular environment containing cytoskeletal networks.
Collapse
Affiliation(s)
- Alexander A Choi
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - Coral Y Zhou
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Ayana Tabo
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, CA 94720
| |
Collapse
|
161
|
da Rocha W, Liberti L, Mucherino A, Malliavin TE. Influence of Stereochemistry in a Local Approach for Calculating Protein Conformations. J Chem Inf Model 2024; 64:8999-9008. [PMID: 39560315 DOI: 10.1021/acs.jcim.4c01232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Protein structure prediction is generally based on the use of local conformational information coupled with long-range distance restraints. Such restraints can be derived from the knowledge of a template structure or the analysis of protein sequence alignment in the framework of models arising from the physics of disordered systems. The accuracy of approaches based on sequence alignment, however, is limited in the case where the number of aligned sequences is small. Here, we derive protein conformations using only local conformations knowledge by means of the interval Branch-and-Prune algorithm. The computation efficiency is directly related to the knowledge of stereochemistry (bond angle and ω values) along the protein sequence and, in particular, to the variations of the torsion angle ω. The impact of stereochemistry variations is particularly strong in the case of protein topologies defined from numerous long-range restraints, as in the case of protein of β secondary structures. The systematic enumeration of the conformations improves the efficiency of the calculations. The analysis of DNA codons permits to connect the variations of torsion angle ω to the positions of rare DNA codons.
Collapse
Affiliation(s)
- Wagner da Rocha
- LIX CNRS, École Polytechnique, Institut Polytechnique de Paris, Palaiseau 91128, France
| | - Leo Liberti
- LIX CNRS, École Polytechnique, Institut Polytechnique de Paris, Palaiseau 91128, France
| | | | - Thérèse E Malliavin
- LPCT, UMR 7019 Université de Lorraine CNRS, Vandoeuvre-lès-Nancy 54500, France
| |
Collapse
|
162
|
Madern D, Halgand F, Houée-Levin C, Dufour AB, Coquille S, Ansanay-Alex S, Sacquin-Mora S, Brochier-Armanet C. The Characterization of Ancient Methanococcales Malate Dehydrogenases Reveals That Strong Thermal Stability Prevents Unfolding Under Intense γ-Irradiation. Mol Biol Evol 2024; 41:msae231. [PMID: 39494471 PMCID: PMC11631191 DOI: 10.1093/molbev/msae231] [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: 04/10/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Malate dehydrogenases (MalDHs) (EC.1.1.1.37), which are involved in the conversion of oxaloacetate to pyruvate in the tricarboxylic acid cycle, are a relevant model for the study of enzyme evolution and adaptation. Likewise, a recent study showed that Methanococcales, a major lineage of Archaea, is a good model to study the molecular processes of proteome thermoadaptation in prokaryotes. Here, we use ancestral sequence reconstruction and paleoenzymology to characterize both ancient and extant MalDHs. We observe a good correlation between inferred optimal growth temperatures and experimental optimal temperatures for activity (A-Topt). In particular, we show that the MalDH present in the ancestor of Methanococcales was hyperthermostable and had an A-Topt of 80 °C, consistent with a hyperthermophilic lifestyle. This ancestor gave rise to two lineages with different thermal constraints: one remained hyperthermophilic, while the other underwent several independent adaptations to colder environments. Surprisingly, the enzymes of the first lineage have retained a thermoresistant behavior (i.e. strong thermostability and high A-Topt), whereas the ancestor of the second lineage shows a strong thermostability, but a reduced A-Topt. Using mutants, we mimic the adaptation trajectory toward mesophily and show that it is possible to significantly reduce the A-Topt without altering the thermostability of the enzyme by introducing a few mutations. Finally, we reveal an unexpected link between thermostability and the ability to resist γ-irradiation-induced unfolding.
Collapse
Affiliation(s)
| | - Frédéric Halgand
- Institut de Chimie Physique, Université Paris-Saclay, 91405 Orsay, France
| | | | - Anne-Béatrice Dufour
- Universite Claude Bernard Lyon 1, LBBE, UMR 5558, CNRS, VAS, Villeurbanne F-69622, France
| | | | | | - Sophie Sacquin-Mora
- Laboratoire de Biochimie Théorique, CNRS, UPR9080, Université Paris-Cité, 75005 Paris, France
- Institut de Biologie Physico-Chimique-Fondation Edmond de Rothschild, PSL Research University, Paris, France
| | - Céline Brochier-Armanet
- Universite Claude Bernard Lyon 1, LBBE, UMR 5558, CNRS, VAS, Villeurbanne F-69622, France
- Institut Universitaire de France (IUF), France
| |
Collapse
|
163
|
Borusak S, Denger K, Dorendorf T, Fournier C, Lerner H, Mayans O, Spiteller D, Schleheck D. Anaerobic Faecalicatena spp. degrade sulfoquinovose via a bifurcated 6-deoxy-6-sulfofructose transketolase/transaldolase pathway to both C 2- and C 3-sulfonate intermediates. Front Microbiol 2024; 15:1491101. [PMID: 39712897 PMCID: PMC11659671 DOI: 10.3389/fmicb.2024.1491101] [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: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 12/24/2024] Open
Abstract
Plant-produced sulfoquinovose (SQ, 6-deoxy-6-sulfoglucose) is one of the most abundant sulfur-containing compounds in nature and its bacterial degradation plays an important role in the biogeochemical sulfur and carbon cycles and in all habitats where SQ is produced and degraded, particularly in gut microbiomes. Here, we report the enrichment and characterization of a strictly anaerobic SQ-degrading bacterial consortium that produces the C2-sulfonate isethionate (ISE) as the major product but also the C3-sulfonate 2,3-dihydroxypropanesulfonate (DHPS), with concomitant production of acetate and hydrogen (H2). In the second step, the ISE was degraded completely to hydrogen sulfide (H2S) when an additional electron donor (external H2) was supplied to the consortium. Through growth experiments, analytical chemistry, genomics, proteomics, and transcriptomics, we found evidence for a combination of the 6-deoxy-6-sulfofructose (SF) transketolase (sulfo-TK) and SF transaldolase (sulfo-TAL) pathways in a SQ-degrading Faecalicatena-phylotype (family Lachnospiraceae) of the consortium, and for the ISE-desulfonating glycyl-radical enzyme pathway, as described for Bilophila wadsworthia, in an Anaerospora-phylotype (Sporomusaceae). Furthermore, using total proteomics, a new gene cluster for a bifurcated SQ pathway was also detected in Faecalicatena sp. DSM22707, which grew with SQ in pure culture, producing mainly ISE, but also 3-sulfolacate (SL) 3-sulfolacaldehyde (SLA), acetate, butyrate, succinate, and formate, but not H2. We then reproduced the growth of the consortium with SQ in a defined co-culture model consisting of Faecalicatena sp. DSM22707 and Bilophila wadsworthia 3.1.6. Our findings provide the first description of an additional sulfoglycolytic, bifurcated SQ pathway. Furthermore, we expand on the knowledge of sulfidogenic SQ degradation by strictly anaerobic co-cultures, comprising SQ-fermenting bacteria and cross-feeding of the sulfonate intermediate to H2S-producing organisms, a process in gut microbiomes that is relevant for human health and disease.
Collapse
Affiliation(s)
- Sabrina Borusak
- Department of Biology, University of Konstanz, Konstanz, Germany
- The Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
- Department of Biology, Limnological Institute, University of Konstanz, Konstanz, Germany
| | - Karin Denger
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Till Dorendorf
- Department of Biology, University of Konstanz, Konstanz, Germany
- The Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Corentin Fournier
- Department of Biology, University of Konstanz, Konstanz, Germany
- Department of Biology, Limnological Institute, University of Konstanz, Konstanz, Germany
| | - Harry Lerner
- Department of Biology, University of Konstanz, Konstanz, Germany
- Department of Biology, Limnological Institute, University of Konstanz, Konstanz, Germany
| | - Olga Mayans
- Department of Biology, University of Konstanz, Konstanz, Germany
- The Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Dieter Spiteller
- Department of Biology, University of Konstanz, Konstanz, Germany
- The Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - David Schleheck
- Department of Biology, University of Konstanz, Konstanz, Germany
- The Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
- Department of Biology, Limnological Institute, University of Konstanz, Konstanz, Germany
| |
Collapse
|
164
|
Fan J, Tan Z, He S, Li A, Jia Y, Li J, Zhang Z, Li B, Chu H. TBAJ-587, a novel diarylquinoline, is active against Mycobacterium abscessus. Antimicrob Agents Chemother 2024; 68:e0094524. [PMID: 39470202 PMCID: PMC11619292 DOI: 10.1128/aac.00945-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
Nontuberculous mycobacteria (NTM) infections are extremely difficult to treat due to a natural resistance to many antimicrobials. TBAJ-587 is a novel diarylquinoline, which shows higher anti-tuberculosis activity, lower lipophilicity, and weaker inhibition of hERG channels than bedaquiline (BDQ). The susceptibilities of 11 NTM reference strains and 194 clinical Mycobacterium abscessus isolates to TBAJ-587 were determined by the broth microdilution assay. The activity of TBAJ-587 toward the growth of M. abscessus in macrophages was also evaluated. Minimum bactericidal concentration and time-kill kinetic assays were conducted to distinguish between the bactericidal and bacteriostatic activities of TBAJ-587. The synergy between TBAJ-587 and eight clinically important antibiotics was determined using a checkerboard assay. TBAJ-587 was highly effective against M. abscessus by targeting its F-ATP synthase c chain. The antimicrobial activities of TBAJ-587 and BDQ toward intracellular M. abscessus were comparable. The in vivo activities of TBAJ-587 and BDQ in an immunocompromised mouse model were also comparable. TBAJ-587 expressed bactericidal activity and was compatible with eight anti-NTM drugs commonly used in clinical practice; no antagonism was discovered. As such, TBAJ-587 represents a potential candidate for the treatment of NTM infections.
Collapse
Affiliation(s)
- Junsheng Fan
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Zhili Tan
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Siyuan He
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Anqi Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Yaping Jia
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Juan Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Zhemin Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Haiqing Chu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
165
|
Rawat K, Tewari D, Bisht A, Chandra S, Tiruneh YK, Hassan HM, Al-Emam A, Sindi ER, Al-Dies AAM. Identification of AChE targeted therapeutic compounds for Alzheimer's disease: an in-silico study with DFT integration. Sci Rep 2024; 14:30356. [PMID: 39638823 PMCID: PMC11621528 DOI: 10.1038/s41598-024-81285-2] [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: 09/20/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition marked by cognitive deterioration and changes in behavior. Acetylcholinesterase (AChE), which hydrolyzes acetylcholine, is a key drug target for treating AD. This research aimed to identify new AChE inhibitors using the IMPPAT database. We used known drugs as a basis to search for similar chemicals in the IMPPAT database and created a library of 127 plant-based compounds. Initial screening of these compounds was performed using molecular docking, followed by an analysis of their drug-likeness and ADMET properties. Compounds with favorable properties underwent density functional theory (DFT) calculations to assess their electronic properties such as HOMO-LUMO gap, electron density, and molecular orbital distribution. These descriptors provided insights into each compound's reactivity, stability, and binding potential with AChE. Promising candidates were further evaluated through molecular dynamics (MD) simulations over 100 ns and MMPBSA analysis for the last 30 ns. Two compounds, Biflavanone (IMPHY013027) with a binding free energy of - 130.394 kcal/mol and Calomelanol J (IMPHY007737) with - 107.908 kcal/mol, demonstrated strong binding affinities compared to the reference molecule HOR, which has a binding free energy of - 105.132 kcal/mol. These compounds exhibited promising drug-ability profiles in both molecular docking and MD simulations, indicating their potential as novel AChE inhibitors for AD treatment. However, further experimental validation is necessary to verify their effectiveness and safety.
Collapse
Affiliation(s)
- Kalpana Rawat
- Computational Biology and Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Disha Tewari
- Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Amisha Bisht
- Department of Botany, Soban Singh Jeena University, Pt. Badridutt Pandey Campus Bageshwar, Almora, Uttarakhand, 263601, India
| | - Subhash Chandra
- Computational Biology and Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India.
| | - Yewulsew Kebede Tiruneh
- Department of Biology, Biomedical Sciences stream, Bahir Dar University, P.O.Box=79, Bahir, Ethiopia.
| | - Hesham M Hassan
- Department of Pathology, College of Medicine, King Khalid University, 61421, Asir, Saudi Arabia
- Department of pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, 61421, Asir, Saudi Arabia
| | - Emad Rashad Sindi
- Division of Clinical Biochemistry, Department of Basic Medical Sciences, College of Medicine, University of Jeddah, 23890, Jeddah, Saudi Arabia
| | - Al-Anood M Al-Dies
- Chemistry Department, Umm Al-Qura University, Al-Qunfudah University College, Mecca, Saudi Arabia
| |
Collapse
|
166
|
Vergara S, Zhou X, Santiago U, Alaoui-El-Azher M, Conway JF, Sluis-Cremer N, Calero G. Structural basis of deoxynucleotide addition by HIV-1 RT during reverse transcription. Nat Commun 2024; 15:10553. [PMID: 39632888 PMCID: PMC11618517 DOI: 10.1038/s41467-024-54618-y] [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: 03/07/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Reverse transcription of the retroviral RNA genome into DNA is an integral step during HIV-1 replication. Despite a wealth of structural information on reverse transcriptase (RT), we lack insight into the intermediate states of DNA synthesis. Using catalytically active substrates, and a blot/diffusion cryo-electron microscopy approach, we capture 11 structures encompassing reactant, intermediate and product states of dATP addition by RT at 2.2 to 3.0 Å resolution. In the reactant state, dATP binding to RT-template/primer involves a single Mg2+ (site B) inducing formation of a negatively charged pocket where a second floating Mg2+ can bind (site A). During the intermediate state, the α-phosphate oxygen from a previously unobserved dATP conformer aligns with site A Mg2+ and the primer 3'-OH for nucleophilic attack. The product state, comprises two substrate conformations including an incorporated dAMP with the pyrophosphate leaving group coordinated by metal B and stabilized through H-bonds. Moreover, K220 mutants significantly impact the rate of dNTP incorporation by RT and HIV-1 replication capacity. This work sheds light into the dynamic components of a reaction that is central to HIV-1 replication.
Collapse
Affiliation(s)
- Sandra Vergara
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaohong Zhou
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ulises Santiago
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mounia Alaoui-El-Azher
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James F Conway
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nicolas Sluis-Cremer
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Guillermo Calero
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
167
|
Pikoulas A, Morianos I, Nidris VN, Hamdy R, López-López A, Moran-Garrido M, Muthu V, Halabalaki M, Papadovasilaki M, Irene K, Gu Y, Aerts R, Mercier T, Vanbiervliet Y, Cho SY, Spallone A, Samonakis D, Kastritis E, Drakos E, Tzardi M, Eliopoulos A, Georgila K, Carvalho A, Kurzai O, Rudramurthy S, Lanternier F, Petratos K, Maertens J, Bruno V, Kontoyiannis D, Barbas C, Soliman S, Ibrahim A, Chamilos G. Albumin orchestrates a natural host defense mechanism against mucormycosis. RESEARCH SQUARE 2024:rs.3.rs-5441197. [PMID: 39678331 PMCID: PMC11643317 DOI: 10.21203/rs.3.rs-5441197/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Mucormycosis is an emerging, life-threatening human infection caused by fungi of the order Mucorales. Metabolic disorders uniquely predispose an ever-expanding group of patients to mucormycosis via poorly understood mechanisms. Therefore, it is highly likely that uncharacterized host metabolic effectors confer protective immunity against mucormycosis. Here, we uncover a master regulatory role of albumin in host defense against Mucorales through the modulation of the fungal pathogenicity program. Our initial studies identified severe hypoalbuminemia as a prominent metabolic abnormality and a biomarker of poor outcome in independent cohorts of mucormycosis patients. Strikingly, we found that purified albumin selectively inhibits Mucorales growth among a range of human pathogens, and albumin-deficient mice display susceptibility specifically to mucormycosis. The antifungal activity of albumin is mediated by the release of bound free fatty acids (FFAs). Importantly, albumin prevents FFA oxidation, which results in loss of their antifungal properties. A high degree of FFA oxidation is found in the sera of patients with mucormycosis. Physiologically, albumin-bound FFAs blocks the expression of the mycotoxin mucoricin and renders Mucorales avirulent in vivo. Overall, we discovered a novel host defense mechanism that directs the pathogen to suppress its growth and the expression of virulence factors in response to unfavorable metabolic cues regulated by albumin. These findings have major implications for the pathogenesis and management of mucormycosis.
Collapse
Affiliation(s)
| | - Ioannis Morianos
- IMBB, FORTH, Nikolaou Plastira 100 GR-70013, Heraklion, Crete GREECE
| | | | - Rania Hamdy
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Angeles López-López
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Ma
| | - Maria Moran-Garrido
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Ma
| | - Valliappan Muthu
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Maria Halabalaki
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Yiyou Gu
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center
| | - Robina Aerts
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Toine Mercier
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Yuri Vanbiervliet
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Sung-Yeon Cho
- Department of Infectious Diseases, Infection Control and Employee Health, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amy Spallone
- Department of Infectious Diseases, Infection Control and Employee Health, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Efstathios Kastritis
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Tzardi
- School of Medicine, University of Crete and University Hospital
| | - Aristides Eliopoulos
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Georgila
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Oliver Kurzai
- Institute for Hygiene and Microbiology, University of Würzburg, 97080, Würzburg, Germany
| | - Shivaprakash Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Fanny Lanternier
- Service de Maladies Infectieuses et Tropicales, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Université Paris Cite, France
| | | | - Johan Maertens
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Universidad San Pablo-CEU
| | | | - Ashraf Ibrahim
- The Lundquist Institute at Harbor-University of California Los Angeles Medical Center
| | | |
Collapse
|
168
|
Ubilla-Rodriguez NC, Andreas MP, Giessen TW. Structural and biochemical characterization of a widespread enterobacterial peroxidase encapsulin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625667. [PMID: 39651212 PMCID: PMC11623594 DOI: 10.1101/2024.11.27.625667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Encapsulins are self-assembling protein compartments found in prokaryotes and specifically encapsulate dedicated cargo enzymes. The most abundant encapsulin cargo class are Dye-decolorizing Peroxidases (DyPs). It has been previously suggested that DyP encapsulins are involved in oxidative stress resistance and bacterial pathogenicity due to DyPs' inherent ability to reduce and detoxify hydrogen peroxide while oxidizing a broad range of organic co-substrates. Here, we report the structural and biochemical analysis of a DyP encapsulin widely found across enterobacteria. Using bioinformatic approaches, we show that this DyP encapsulin is encoded by a conserved transposon-associated operon, enriched in enterobacterial pathogens. Through low pH and peroxide exposure experiments, we highlight the stability of this DyP encapsulin under harsh conditions and show that DyP catalytic activity is highest at low pH. We determine the structure of the DyP-loaded shell and free DyP via cryo-electron microscopy, revealing the structural basis for DyP cargo loading and peroxide preference. Our work lays the foundation to further explore the substrate range and physiological functions of enterobacterial DyP encapsulins.
Collapse
|
169
|
Caetano-Anollés G, Mughal F, Aziz MF, Caetano-Anollés K. Tracing the birth and intrinsic disorder of loops and domains in protein evolution. Biophys Rev 2024; 16:723-735. [PMID: 39830125 PMCID: PMC11735766 DOI: 10.1007/s12551-024-01251-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025] Open
Abstract
Protein loops and structural domains are building blocks of molecular structure. They hold evolutionary memory and are largely responsible for the many functions and processes that drive the living world. Here, we briefly review two decades of phylogenomic data-driven research focusing on the emergence and evolution of these elemental architects of protein structure. Phylogenetic trees of domains reconstructed from the proteomes of organisms belonging to all three superkingdoms and viruses were used to build chronological timelines describing the origin of each domain and its embedded loops at different levels of structural abstraction. These timelines consistently recovered six distinct evolutionary phases and a most parsimonious evolutionary progression of cellular life. The timelines also traced the birth of domain structures from loops, which allowed to model their growth ab initio with AlphaFold2. Accretion decreased the disorder of the growing molecules, suggesting disorder is molecular size-dependent. A phylogenomic survey of disorder revealed that loops and domains evolved differently. Loops were highly disordered, disorder increased early in evolution, and ordered and moderate disordered structures were derived. Gradual replacement of loops with α-helix and β-strand bracing structures over time paved the way for the dominance of more disordered loop types. In contrast, ancient domains were ordered, with disorder evolving as a benefit acquired later in evolution. These evolutionary patterns explain inverse correlations between disorder and sequence length of loops and domains. Our findings provide a deep evolutionary view of the link between structure, disorder, flexibility, and function.
Collapse
Affiliation(s)
- Gustavo Caetano-Anollés
- Evolutionary Bioinformatics Laboratory, Department of Crop Sciences and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Fizza Mughal
- Evolutionary Bioinformatics Laboratory, Department of Crop Sciences and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - M. Fayez Aziz
- Evolutionary Bioinformatics Laboratory, Department of Crop Sciences and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Kelsey Caetano-Anollés
- Evolutionary Bioinformatics Laboratory, Department of Crop Sciences and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Callout Biotech, Albuquerque, NM 87112 USA
| |
Collapse
|
170
|
Shankar G, Akhter Y. Stealing survival: Iron acquisition strategies of Mycobacteriumtuberculosis. Biochimie 2024; 227:37-60. [PMID: 38901792 DOI: 10.1016/j.biochi.2024.06.006] [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: 05/06/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), faces iron scarcity within the host due to immune defenses. This review explores the importance of iron for Mtb and its strategies to overcome iron restriction. We discuss how the host limits iron as an innate immune response and how Mtb utilizes various iron acquisition systems, particularly the siderophore-mediated pathway. The review illustrates the structure and biosynthesis of mycobactin, a key siderophore in Mtb, and the regulation of its production. We explore the potential of targeting siderophore biosynthesis and uptake as a novel therapeutic approach for TB. Finally, we summarize current knowledge on Mtb's iron acquisition and highlight promising directions for future research to exploit this pathway for developing new TB interventions.
Collapse
Affiliation(s)
- Gauri Shankar
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India.
| |
Collapse
|
171
|
Haldiya A, Kain H, Dubey S, Punde SK, Gupta PKP, Srivastava VK, Srivastava SK, Kothari SL, Kaushik S. Investigating Sortase A inhibitory potential of herbal compounds using integrated computational and biochemical approaches. Acta Trop 2024; 260:107430. [PMID: 39413895 DOI: 10.1016/j.actatropica.2024.107430] [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: 08/31/2024] [Revised: 10/06/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Multi-drug resistance in bacteria is emerging as a major global health challenge, causing substantial harm in terms of mortality, morbidity, and financial strain on healthcare systems. These bacteria are constantly acquiring new virulence factors and drug-resistance mechanisms, which highlights the critical need for innovative antimicrobial medicines and identification of new therapeutic targets, such as Sortase A (EfSrtAΔN59). EfSrtAΔN59, a transpeptidase significant for the adhesion and virulence of Enterococcus faecalis (E. faecalis), presents an attractive target for disrupting biofilm formation-a key factor in persistent infections. This study investigates the inhibitory effects of two natural flavonoids- Rutin Trihydrate and Quercetin, on EfSrtAΔN59 and biofilm formation in E. faecalis. With in vitro enzymatic assays and biofilm quantification techniques, we demonstrate that both compounds significantly attenuate EfSrtAΔN59 activity, thereby hindering bacterial biofilm formation. Rutin Trihydrate and Quercetin exhibited strong binding affinities to the EfSrtAΔN59 enzyme, as confirmed by molecular docking and MD simulation studies. This was further substantiated by a notable reduction in biofilm biomass in bacterial cultures treated with these compounds. These findings highlight the potential of Rutin Trihydrate and Quercetin as promising candidates for the development of novel anti-virulence therapies aimed at mitigating E. faecalis infections, thereby offering a compelling alternative to traditional antibiotics.
Collapse
Affiliation(s)
- Akanksha Haldiya
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan 303002, India
| | - Himanshi Kain
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan 303002, India
| | - Saumya Dubey
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Off Jaipur-Ajmer Expressway, Jaipur, Rajasthan 303007, India
| | | | - Pramod Kumar P Gupta
- School of Biotechnology and Bioinformatics, D Y Patil Deemed to be University, Plot 50 Sector 15 CBD Belapur, Navi Mumbai, Maharashtra 400614, India
| | | | - Sandeep Kumar Srivastava
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Off Jaipur-Ajmer Expressway, Jaipur, Rajasthan 303007, India
| | - S L Kothari
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan 303002, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan 303002, India.
| |
Collapse
|
172
|
Yang Z, Zhong W, Lv Q, Dong T, Chen G, Chen CYC. Interaction-Based Inductive Bias in Graph Neural Networks: Enhancing Protein-Ligand Binding Affinity Predictions From 3D Structures. IEEE TRANSACTIONS ON PATTERN ANALYSIS AND MACHINE INTELLIGENCE 2024; 46:8191-8208. [PMID: 38739515 DOI: 10.1109/tpami.2024.3400515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Inductive bias in machine learning (ML) is the set of assumptions describing how a model makes predictions. Different ML-based methods for protein-ligand binding affinity (PLA) prediction have different inductive biases, leading to different levels of generalization capability and interpretability. Intuitively, the inductive bias of an ML-based model for PLA prediction should fit in with biological mechanisms relevant for binding to achieve good predictions with meaningful reasons. To this end, we propose an interaction-based inductive bias to restrict neural networks to functions relevant for binding with two assumptions: 1) A protein-ligand complex can be naturally expressed as a heterogeneous graph with covalent and non-covalent interactions; 2) The predicted PLA is the sum of pairwise atom-atom affinities determined by non-covalent interactions. The interaction-based inductive bias is embodied by an explainable heterogeneous interaction graph neural network (EHIGN) for explicitly modeling pairwise atom-atom interactions to predict PLA from 3D structures. Extensive experiments demonstrate that EHIGN achieves better generalization capability than other state-of-the-art ML-based baselines in PLA prediction and structure-based virtual screening. More importantly, comprehensive analyses of distance-affinity, pose-affinity, and substructure-affinity relations suggest that the interaction-based inductive bias can guide the model to learn atomic interactions that are consistent with physical reality. As a case study to demonstrate practical usefulness, our method is tested for predicting the efficacy of Nirmatrelvir against SARS-CoV-2 variants. EHIGN successfully recognizes the changes in the efficacy of Nirmatrelvir for different SARS-CoV-2 variants with meaningful reasons.
Collapse
|
173
|
de Oliveira Santos LAB, Batista MVDA. Structure-based virtual screening and drug repurposing studies indicate potential inhibitors of bovine papillomavirus E6 oncoprotein. Microbiol Immunol 2024; 68:414-426. [PMID: 39467039 DOI: 10.1111/1348-0421.13178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/24/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
Bovine papillomavirus type 1 (BPV1) is an oncogenic virus that causes lesions and cancer in infected cattle. Despite being one of the most studied genotypes in the family and occurring in herds worldwide, there are currently no vaccines or drugs for its control. The viral E6 oncoprotein plays a crucial role in infection by this virus, making it a promising target for the development of new therapies. In this regard, we integrated structure-based virtual screening approaches, drug repositioning, and molecular dynamics to identify approved drugs with the potential to inhibit BPV1 E6. Our results reveal that Lumacaftor and MK-3207 are promising candidates for controlling BPV1 infection. The findings of this study may contribute to the development of E6 oncoprotein blockers in an accelerated and cost-effective manner.
Collapse
Affiliation(s)
- Lucas Alexandre Barbosa de Oliveira Santos
- Laboratory of Molecular Genetics and Biotechnology, Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology, Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| |
Collapse
|
174
|
Kannan A, Naganathan AN. Engineering the native ensemble to tune protein function: Diverse mutational strategies and interlinked molecular mechanisms. Curr Opin Struct Biol 2024; 89:102940. [PMID: 39393291 DOI: 10.1016/j.sbi.2024.102940] [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: 07/30/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
Natural proteins are fragile entities, intrinsically sensitive to perturbations both at the level of sequence and their immediate environment. Here, we highlight the diverse strategies available for engineering function through mutations influencing backbone conformational entropy, charge-charge interactions, and in the loops and hinge regions, many of which are located far from the active site. It thus appears that there are potentially numerous ways to microscopically vary the identity of residues and the constituent interactions to tune function. Functional modulation could occur via changes in native-state stability, altered thermodynamic coupling extents within the folded structure, redistributed dynamics, or through modulation of the population of conformational substates. As these mechanisms are intrinsically linked and given the pervasive long-range effects of mutations, it is crucial to consider the interaction network as a whole and fully map the native conformational landscape to place mutational effects in the context of allostery and protein evolution.
Collapse
Affiliation(s)
- Adithi Kannan
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Athi N Naganathan
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
175
|
Elgushe SM, El-Sonbati AZ, Diab MA, Gomaa EA, AbouElleef EM. Eugenol's electrochemical behavior, complexation interaction with copper chloride, antioxidant activity, and potential drug molecular docking application for Covid-19. Colloids Surf B Biointerfaces 2024; 244:114194. [PMID: 39226846 DOI: 10.1016/j.colsurfb.2024.114194] [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: 05/11/2024] [Revised: 06/19/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
Electrochemical studies were conducted to analyze the behavior of eugenol, CuCl2, and their complex using cyclic voltammetry. The oxidation mechanisms of eugenol and the redox behavior of copper ions were elucidated, showing differences in reversibility and charge transfer coefficients. Various kinetic and solvation parameters were determined. The redox behavior of CuCl2 was found to be more reversible compared to the copper-eugenol complex. The copper-eugenol complex exhibited enhanced antioxidant activity compared to eugenol and standard ascorbic acid. The eugenol was oxidized to form eugenol quinone methide through two postulated irreversible mechanisms. Molecular docking studies suggested higher potential bioactivity of the copper-eugenol complex towards the target protein of COVID-19 than the eugenol ligand.
Collapse
Affiliation(s)
- Saleh M Elgushe
- Chemistry Department, Faculty of Science, Damietta University, Egypt
| | - Adel Z El-Sonbati
- Chemistry Department, Faculty of Science, Damietta University, Egypt
| | - Mostafa A Diab
- Chemistry Department, Faculty of Science, Damietta University, Egypt
| | - Esam A Gomaa
- Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Elsayed M AbouElleef
- Basic Sciences Department, Delta Higher Institute for Engineering and Technology, Mansoura, Dakhlia 35681, Egypt.
| |
Collapse
|
176
|
Caffrey BJ, Pedrazo‐Tardajos A, Liberti E, Gaunt B, Kim JS, Kirkland AI. Liquid Phase Electron Microscopy of Bacterial Ultrastructure. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402871. [PMID: 39239997 PMCID: PMC11636060 DOI: 10.1002/smll.202402871] [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: 04/10/2024] [Revised: 06/05/2024] [Indexed: 09/07/2024]
Abstract
Recent advances in liquid phase scanning transmission electron microscopy (LP-STEM) have enabled the study of dynamic biological processes at nanometer resolutions, paving the way for live-cell imaging using electron microscopy. However, this technique is often hampered by the inherent thickness of whole cell samples and damage from electron beam irradiation. These restrictions degrade image quality and resolution, impeding biological interpretation. Using graphene encapsulation, scanning transmission electron microscopy (STEM), and energy-dispersive X-ray (EDX) spectroscopy to mitigate these issues provides unprecedented levels of intracellular detail in aqueous specimens. This study demonstrates the potential of LP-STEM to examine and identify internal cellular structures in thick biological samples. Specifically, it highlights the use of LP-STEM to investigate the radiation resistant, gram-positive bacterium, Deinococcus radiodurans using various imaging techniques.
Collapse
Affiliation(s)
- Brian J. Caffrey
- The Rosalind Franklin InstituteHarwell Science and Innovation CampusDidcotOX11 OQXUK
| | | | - Emanuela Liberti
- The Rosalind Franklin InstituteHarwell Science and Innovation CampusDidcotOX11 OQXUK
| | - Benjamin Gaunt
- The Rosalind Franklin InstituteHarwell Science and Innovation CampusDidcotOX11 OQXUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordJohn Radcliffe HospitalOxfordOX3 9DUUK
| | - Judy S. Kim
- The Rosalind Franklin InstituteHarwell Science and Innovation CampusDidcotOX11 OQXUK
- Department of MaterialsUniversity of OxfordOxfordOX1 3PHUK
| | - Angus I. Kirkland
- The Rosalind Franklin InstituteHarwell Science and Innovation CampusDidcotOX11 OQXUK
- Department of MaterialsUniversity of OxfordOxfordOX1 3PHUK
| |
Collapse
|
177
|
Erdogan MK, Sever A, Gundogdu R, Toy Y, Gecibesler IH, Yapar Y, Behcet L, Zengin G. Verbascum gimgimense an Endemic Turkish Plant: Evaluation of In Vitro Anticancer, Antioxidant, Enzyme Inhibitory Activities, and Phytochemical Profile. Cell Biochem Funct 2024; 42:e70023. [PMID: 39632482 DOI: 10.1002/cbf.70023] [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: 08/23/2024] [Revised: 10/21/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
The Verbascum genus has gained significant attention in the pharmaceutical field, particularly in recent years, due to its valuable medicinal properties, which are well-recognized in complementary and alternative medicine. Certain species within this genus contain essential compounds and exhibit a wide range of therapeutic activities. In this study, the ethanolic extract of Verbascum gimgimense (VG) was analyzed for its cytotoxic, apoptotic, antioxidant, and enzyme inhibitory properties, as well as its phenolic and lipophilic compounds. The phenolic compounds in the extract were identified using Exactive Plus Orbitrap HPLC-HRMS, while the lipophilic components were characterized by GC-MS analysis. The Neutral Red Uptake (NRU) cell viability assay and colony formation assay were performed to assess the antiproliferative and anti-colony survival effects of VG on the A549 human lung adenocarcinoma cell line. Additionally, a wound healing assay measured cell migration, and the apoptotic process was evaluated using Caspase-3 ELISA and acridine orange/ethidium bromide staining. Protein expression levels were determined by western blot analysis. DPPH, ABTS FRAP, and CUPRAC assays were used to determine free radical scavenging, reducing power, and metal chelating activities, respectively. VG was rich in dominant phenolic components, including benzoic acid (6.809 mg/g extract), phloretic acid (1.279 mg/g extract), luteolin 7-rutinoside (2.799 mg/g extract), luteoloside (3.300 mg/g extract), kuromanine (3.456 mg/g extract), and rutin hydrate (2.015 mg/g extract). Major fatty acids identified in VG included palmitic acid (17.3%), stearic acid (2.99%), linoleic acid (9.44%), and α-linolenic acid (26.48%). VG treatment significantly reduced colony formation ability, decreased wound closure, and increased both apoptotic cell count and caspase-3 activity compared to the control group. Protein levels of c-PARP, p53, and p21 were substantially elevated compared to controls. In addition to its strong free radical scavenging, reducing power and metal chelating activity, VG exhibited strong inhibitory effects on α-amylase, α-glucosidase, AChE, BChE, and tyrosinase. Our study demonstrates that VG possesses antiproliferative, apoptotic, antioxidant, and enzyme-inhibitory properties. V. gimgimense emerges as a promising natural antioxidant source with potentially significant regulatory effects on key enzymes and proteins, which could contribute to managing various human diseases and inspire the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Aydın Sever
- Department of Pharmacy Services, Vocational School of Health Services, Bingol University, Bingol, Türkiye
| | - Ramazan Gundogdu
- Department of Pharmacy Services, Vocational School of Health Services, Bingol University, Bingol, Türkiye
| | - Yusuf Toy
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Ibrahim Halil Gecibesler
- Department of Occupational Health and Safety, Faculty of Health Science, Bingol University, Bingol, Türkiye
| | - Yakup Yapar
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Lutfi Behcet
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Türkiye
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Türkiye
| |
Collapse
|
178
|
Pierson JA, Yang JE, Wright ER. Recent advances in correlative cryo-light and electron microscopy. Curr Opin Struct Biol 2024; 89:102934. [PMID: 39366119 PMCID: PMC11602379 DOI: 10.1016/j.sbi.2024.102934] [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: 05/21/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024]
Abstract
Correlative light and electron microscopy (CLEM) pipelines serve to integrate the imaging modalities of fluorescence light microscopy (FLM) and cryogenic electron microscopy (cryo-EM) to produce contextually relevant high-resolution structural snapshots of biological systems. Innovations in sample preparation, instrumentation, imaging, and data processing have advanced the field of cryo-EM. This review focuses on prior work and recent developments in the field of cryo- EM that support further integration of technologies for correlative microscopy workflows.
Collapse
Affiliation(s)
- Joshua A Pierson
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA
| | - Jie E Yang
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA; Cryo-Electron Microscopy Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, USA; Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, USA
| | - Elizabeth R Wright
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA; Cryo-Electron Microscopy Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, USA; Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA.
| |
Collapse
|
179
|
Liu Z, Grigas AT, Sumner J, Knab E, Davis CM, O'Hern CS. Identifying the minimal sets of distance restraints for FRET-assisted protein structural modeling. Protein Sci 2024; 33:e5219. [PMID: 39548730 PMCID: PMC11568256 DOI: 10.1002/pro.5219] [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] [Accepted: 10/26/2024] [Indexed: 11/18/2024]
Abstract
Proteins naturally occur in crowded cellular environments and interact with other proteins, nucleic acids, and organelles. Since most previous experimental protein structure determination techniques require that proteins occur in idealized, non-physiological environments, the effects of realistic cellular environments on protein structure are largely unexplored. Recently, Förster resonance energy transfer (FRET) has been shown to be an effective experimental method for investigating protein structure in vivo. Inter-residue distances measured in vivo can be incorporated as restraints in molecular dynamics (MD) simulations to model protein structural dynamics in vivo. Since most FRET studies only obtain inter-residue separations for a small number of amino acid pairs, it is important to determine the minimum number of restraints in the MD simulations that are required to achieve a given root-mean-square deviation (RMSD) from the experimental structural ensemble. Further, what is the optimal method for selecting these inter-residue restraints? Here, we implement several methods for selecting the most important FRET pairs and determine the number of pairsN r $$ {N}_r $$ that are needed to induce conformational changes in proteins between two experimentally determined structures. We find that enforcing only a small fraction of restraints,N r / N ≲ 0.08 $$ {N}_r/N\lesssim 0.08 $$ , whereN $$ N $$ is the number of amino acids, can induce the conformational changes. These results establish the efficacy of FRET-assisted MD simulations for atomic scale structural modeling of proteins in vivo.
Collapse
Affiliation(s)
- Zhuoyi Liu
- Department of Mechanical Engineering and Materials ScienceYale UniversityNew HavenConnecticutUSA
- Integrated Graduate Program in Physical and Engineering BiologyYale UniversityNew HavenConnecticutUSA
| | - Alex T. Grigas
- Integrated Graduate Program in Physical and Engineering BiologyYale UniversityNew HavenConnecticutUSA
- Graduate Program in Computational Biology and BioinformaticsYale UniversityNew HavenConnecticutUSA
| | - Jacob Sumner
- Integrated Graduate Program in Physical and Engineering BiologyYale UniversityNew HavenConnecticutUSA
- Graduate Program in Computational Biology and BioinformaticsYale UniversityNew HavenConnecticutUSA
| | - Edward Knab
- Department of ChemistryYale UniversityNew HavenConnecticutUSA
| | | | - Corey S. O'Hern
- Department of Mechanical Engineering and Materials ScienceYale UniversityNew HavenConnecticutUSA
- Integrated Graduate Program in Physical and Engineering BiologyYale UniversityNew HavenConnecticutUSA
- Graduate Program in Computational Biology and BioinformaticsYale UniversityNew HavenConnecticutUSA
- Department of PhysicsYale UniversityNew HavenConnecticutUSA
- Department of Applied PhysicsYale UniversityNew HavenConnecticutUSA
| |
Collapse
|
180
|
van de Weert M, Schönbeck C. Ligand binding to proteins - When flawed fluorescence quenching methodology and interpretation become the new norm. Eur J Pharm Sci 2024; 203:106930. [PMID: 39383973 DOI: 10.1016/j.ejps.2024.106930] [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: 09/06/2024] [Accepted: 10/07/2024] [Indexed: 10/11/2024]
Abstract
Intrinsic protein fluorescence quenching measurements have become a widespread methodology to determine ligand-binding properties of in particular serum albumin. Particularly common is the use of double log equations to extract parameters like binding constant and stoichiometry and/or number of binding sites. In this article we discuss that the methodology has several significant and often unrecognized pitfalls, and the double log equations are improperly derived for their purported use. Using simulations, it is shown that the binding stoichiometry and binding constants obtained using these equations may differ substantially from their true values. In addition, it is illustrated how this methodology, via the use of site markers, is unsuited to determine the binding site of ligands on serum albumin. We thus call for a reassessment of the literature in which this methodology plays a central role in characterizing ligand binding to proteins.
Collapse
Affiliation(s)
- Marco van de Weert
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Ubiversitetsparken 2, Copenhagen 2100, Denmark.
| | - Christian Schönbeck
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Ubiversitetsparken 2, Copenhagen 2100, Denmark
| |
Collapse
|
181
|
Malysheva DO, Dymova MA, Richter VA. Analyzing aptamer structure and interactions: in silico modelling and instrumental methods. Biophys Rev 2024; 16:685-700. [PMID: 39830127 PMCID: PMC11735759 DOI: 10.1007/s12551-024-01252-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/31/2024] [Indexed: 01/22/2025] Open
Abstract
Aptamers are short oligonucleotides that bind specifically to various ligands and are characterized by their low immunogenicity, thermostability, and ease of labeling. Many biomedical applications of aptamers as biosensors and drug delivery agents are currently being actively researched. Selective affinity selection with exponential ligand enrichment (SELEX) allows to discover aptamers for a specific target, but it only provides information about the sequence of aptamers; hence other approaches are used for determining aptamer structure, aptamer-ligand interactions and the mechanism of action. The first one is in silico modelling that allows to infer likely secondary and tertiary structures and model their interactions with a ligand. The second approach is to use instrumental methods to study structure and aptamer-ligand interaction. In silico modelling and instrumental methods are complimentary and their combined use allows to eliminate some ambiguity in their respective results. This review examines both the advantages and limitations of in silico modelling and instrumental approaches currently used to study aptamers, which will allow researchers to develop optimal study designs for analyzing aptamer structure and ligand interactions.
Collapse
Affiliation(s)
- Daria O. Malysheva
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
- Physics Department, Novosibirsk State University, Novosibirsk, Russia
| | - Maya A. Dymova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
| | - Vladimir A. Richter
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
| |
Collapse
|
182
|
Ruff AJ. Food industry side streams: an unexploited source for biotechnological phosphorus upcycling. Curr Opin Biotechnol 2024; 90:103209. [PMID: 39326130 DOI: 10.1016/j.copbio.2024.103209] [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: 07/14/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
The phosphorus shortage is an unavoidable challenge that requires strategies to replace phosphorus sourced from ores. Food industry by-products are an unscoped resource for sustainable phosphorus recovery. Recent advances include biotechnological phosphorus upcycling from phytate-rich plant residues to polyphosphate as a food additive. The valorization of by-products such as deoiled seeds or brans additionally provides low-phosphorus feed and thereby minimizes the environmental burden. Phytate reduction in a cereal-rich diet by adding enzyme formulation is a further strategy that limits its antinutritive effect. However, sustainable P-management depends on phytases that have been customized and enhanced for thermostability and specific activity. The circular phosphorus economy is driven by emerging value chains and maturing phosphorus recovery technologies for market entry.
Collapse
Affiliation(s)
- Anna Joëlle Ruff
- Aachen Biology und Biotechnology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
183
|
Cuthbert BJ, Chen X, Burley K, Batot G, Contreras H, Dixon S, Goulding CW. Structural Characterization of Mycobacterium tuberculosis Encapsulin in Complex with Dye-Decolorizing Peroxide. Microorganisms 2024; 12:2465. [PMID: 39770668 PMCID: PMC11676171 DOI: 10.3390/microorganisms12122465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis, the world's deadliest infectious disease. Mtb uses a variety of mechanisms to evade the human host's defenses and survive intracellularly. Mtb's oxidative stress response enables Mtb to survive within activated macrophages, an environment with reactive oxygen species and low pH. Dye-decolorizing peroxidase (DyP), an enzyme involved in Mtb's oxidative stress response, is encapsulated in a nanocompartment, encapsulin (Enc), and is important for Mtb's survival in macrophages. Encs are homologs of viral capsids and encapsulate cargo proteins of diverse function, including those involved in iron storage and stress responses. DyP contains a targeting peptide (TP) at its C-terminus that recognizes and binds to the interior of the Enc nanocompartment. Here, we present the crystal structure of the Mtb-Enc•DyP complex and compare it to cryogenic-electron microscopy (cryo-EM) Mtb-Enc structures. Investigation into the canonical pores formed at symmetrical interfaces reveals that the five-fold pore for the Mtb-Enc crystal structure is strikingly different from that observed in cryo-EM structures. We also observe DyP-TP electron density within the Mtb-Enc shell. Finally, investigation into crystallographic small-molecule binding sites gives insight into potential novel avenues by which substrates could enter Mtb-Enc to react with Mtb-DyP.
Collapse
Affiliation(s)
- Bonnie J. Cuthbert
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Xiaorui Chen
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Kalistyn Burley
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Gaëlle Batot
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Heidi Contreras
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Shandee Dixon
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Celia W. Goulding
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
184
|
Mitrovic SA, Demalgiriya-Gamage C, Winter LM, Kiechle T, Ebenhoch R, Neubauer H, Stierstorfer B, Frego L, Wolfrum C, Reindl S, Nar H. GPR180 is a new member of the Golgi-dynamics domain seven-transmembrane helix protein family. Commun Biol 2024; 7:1588. [PMID: 39609618 PMCID: PMC11605061 DOI: 10.1038/s42003-024-07260-9] [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: 06/26/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024] Open
Abstract
GOLD domain seven-transmembrane helix (GOST) proteins form a new protein family involved in trafficking of membrane-associated cargo. They share a characteristic extracellular/luminal Golgi-dynamics (GOLD) domain, possibly responsible for ligand recognition. Based on structural homology, GPR180 is a new member of this protein family, but little is known about the cellular role of GPR180. Here we show the X-ray structure of the N-terminal domain of GPR180 (1.9 Å) and can confirm the homology to GOLD domains. Using cellular imaging we show the localization of GPR180 in intracellular vesicular structures implying its exposure to acidic pH environments. With Hydrogen/Deuterium Exchange-Mass Spectrometry (HDX-MS) we identify pH-dependent conformational changes, which can be mapped to a putative ligand binding site in the transmembrane region. The results reveal GPR180's role in intracellular vesicles and offer insights into the pH-dependent function of this conserved GOST protein.
Collapse
Affiliation(s)
- Sarah-Ana Mitrovic
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Lisa-Maria Winter
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Tobias Kiechle
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Rebecca Ebenhoch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Heike Neubauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | | | - Lee Frego
- Boehringer Ingelheim, Ridgefield, CT, USA
| | - Christian Wolfrum
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Sophia Reindl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany.
| | - Herbert Nar
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany.
| |
Collapse
|
185
|
Jabeen A, Oakeshott JG, Lee SF, Ranganathan S, Taylor PW. Template-based modeling of insect odorant receptors outperforms AlphaFold3 for ligand binding predictions. Sci Rep 2024; 14:29084. [PMID: 39580516 PMCID: PMC11585542 DOI: 10.1038/s41598-024-80094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Insects rely on odorant receptors (ORs) to detect and respond to volatile environmental cues, so the ORs are attracting increasing interest as potential targets for pest control. However, experimental analysis of their structures and functions faces significant challenges. Computational methods such as template-based modeling (TBM) and AlphaFold3 (AF3) could facilitate the structural characterisation of ORs. This study first showed that both models accurately predicted the structural fold of MhOR5, a jumping bristletail OR with known experimental 3D structures, although accuracy was higher in the extracellular region of the protein and binding mode of their cognate ligands with TBM. The two approaches were then compared for their ability to predict the empirical binding evidence available for OR-odorant complexes in two economically important fruit fly species, Bactrocera dorsalis and B. minax. Post-simulation analyses including binding affinities, complex and ligand stability and receptor-ligand interactions (RLIs) revealed that TBM performed better than AF3 in discriminating between binder and non-binder complexes. TBM's superior performance is attributed to hydrophobicity-based helix-wise multiple sequence alignment (MSA) between available insect OR templates and the ORs for which the binding data were generated. This MSA identified conserved residues and motifs which could be used as anchor points for refining the alignments.
Collapse
Affiliation(s)
- Amara Jabeen
- Applied BioSciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| | | | - Siu Fai Lee
- Applied BioSciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
- CSIRO Environment, Black Mountain, ACT, Australia
| | - Shoba Ranganathan
- Applied BioSciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Phillip W Taylor
- Applied BioSciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| |
Collapse
|
186
|
Bendall EE, Dimcheff D, Papalambros L, Fitzsimmons WJ, Zhu Y, Schmitz J, Halasa N, Chappell J, Martin ET, Biddle JE, Smith-Jeffcoat SE, Rolfes MA, Mellis A, Talbot HK, Grijalva C, Lauring AS. In depth sequencing of a serially sampled household cohort reveals the within-host dynamics of Omicron SARS-CoV-2 and rare selection of novel spike variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624722. [PMID: 39605326 PMCID: PMC11601520 DOI: 10.1101/2024.11.21.624722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
SARS-CoV-2 has undergone repeated and rapid evolution to circumvent host immunity. However, outside of prolonged infections in immunocompromised hosts, within-host positive selection has rarely been detected. The low diversity within-hosts and strong genetic linkage among genomic sites make accurately detecting positive selection difficult. Longitudinal sampling is a powerful method for detecting selection that has seldom been used for SARS-CoV-2. Here we combine longitudinal sampling with replicate sequencing to increase the accuracy of and lower the threshold for variant calling. We sequenced 577 specimens from 105 individuals from a household cohort primarily during the BA.1/BA.2 variant period. There was extremely low diversity and a low rate of divergence. Specimens had 0-12 intrahost single nucleotide variants (iSNV) at >0.5% frequency, and the majority of the iSNV were at frequencies <2%. Within-host dynamics were dominated by genetic drift and purifying selection. Positive selection was rare but highly concentrated in spike. Two individuals with BA.1 infections had S:371F, a lineage defining substitution for BA.2. A Wright Fisher Approximate Bayesian Computational model identified positive selection at 14 loci with 7 in spike, including S:448 and S:339. We also detected significant genetic hitchhiking between synonymous changes and nonsynonymous iSNV under selection. The detectable immune-mediated selection may be caused by the relatively narrow antibody repertoire in individuals during the early Omicron phase of the SARS-CoV-2 pandemic. As both the virus and population immunity evolve, understanding the corresponding shifts in SARS-CoV-2 within-host dynamics will be important.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Derek Dimcheff
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leigh Papalambros
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan Schmitz
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Natasha Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | - H. Keipp Talbot
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carlos Grijalva
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam S. Lauring
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
187
|
Oshiro RT, Dunham DT, Seed KD. The vibriophage-encoded inhibitor OrbA abrogates BREX-mediated defense through the ATPase BrxC. J Bacteriol 2024; 206:e0020624. [PMID: 39404463 PMCID: PMC11580459 DOI: 10.1128/jb.00206-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Bacteria and phages are locked in a co-evolutionary arms race where each entity evolves mechanisms to restrict the proliferation of the other. Phage-encoded defense inhibitors have proven powerful tools to interrogate how defense systems function. A relatively common defense system is BREX (bacteriophage exclusion); however, how BREX functions to restrict phage infection remains poorly understood. A BREX system encoded by the sulfamethoxazole and trimethoprim (SXT) integrative and conjugative element, VchInd5, was recently identified in Vibrio cholerae, the causative agent of the diarrheal disease cholera. The lytic phage ICP1 (International Centre for Diarrhoeal Disease Research, Bangladesh cholera phage 1) that co-circulates with V. cholerae encodes the BREX-inhibitor OrbA, but how OrbA inhibits BREX is unclear. Here, we determine that OrbA inhibits BREX using a unique mechanism from known BREX inhibitors by directly binding to the BREX component BrxC. BrxC has a functional ATPase domain that, when mutated, not only disrupts BrxC function but also alters how BrxC multimerizes. Furthermore, we find that OrbA binding disrupts BrxC-BrxC interactions. We determine that OrbA cannot bind BrxC encoded by the distantly related BREX system encoded by the aSXT VchBan9, and thus fails to inhibit this BREX system that also circulates in epidemic V. cholerae. Lastly, we find that homologs of the VchInd5 BrxC are more diverse than the homologs of the VchBan9 BrxC. These data provide new insight into the function of the BrxC ATPase and highlight how phage-encoded inhibitors can disrupt phage defense systems using different mechanisms.IMPORTANCEWith renewed interest in phage therapy to combat antibiotic-resistant pathogens, understanding the mechanisms bacteria use to defend themselves against phages and the counter-strategies phages evolve to inhibit defenses is paramount. Bacteriophage exclusion (BREX) is a common defense system with few known inhibitors. Here, we probe how the vibriophage-encoded inhibitor OrbA inhibits the BREX system of Vibrio cholerae, the causative agent of the diarrheal disease cholera. By interrogating OrbA function, we have begun to understand the importance and function of a BREX component. Our results demonstrate the importance of identifying inhibitors against defense systems, as they are powerful tools for dissecting defense activity and can inform strategies to increase the efficacy of some phage therapies.
Collapse
Affiliation(s)
- Reid T. Oshiro
- Department of Plant & Microbial Biology, University of California Berkeley, Berkeley, California, USA
| | - Drew T. Dunham
- Department of Plant & Microbial Biology, University of California Berkeley, Berkeley, California, USA
| | - Kimberley D. Seed
- Department of Plant & Microbial Biology, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
188
|
Ma T, Chu J. Chemical structure metagenomics of microbial natural products: surveying nonribosomal peptides and beyond. Beilstein J Org Chem 2024; 20:3050-3060. [PMID: 39600953 PMCID: PMC11590018 DOI: 10.3762/bjoc.20.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Bioactivity-guided fractionation (BGF) has historically been a fruitful natural product discovery workflow. However, it is plagued by increasing rediscovery rates in recent years and new methods capable of exploring the natural product chemical space more broadly and more efficiently is in urgent need. Chemical structure metagenomics as one such method is the theme of this Perspective. It emphasizes a chemical-structure-centered viewpoint toward natural product research. Key to chemical structure metagenomics is the ability to predict the structure of a natural product based on its biosynthetic gene sequences, which facilitated the discovery of numerous new bioactive molecules and helped uncover oversampled/underexplored niches of decades of BGF based discovery. While microbial nonribosomal peptides have been the focus of chemical structure metagenomics efforts thus far, it is in principle applicable to other natural product families. The future outlook of this new approach will also be discussed.
Collapse
Affiliation(s)
- Thomas Ma
- Department of Chemistry, National Taiwan University, Taipei City 10617, Taiwan
| | - John Chu
- Department of Chemistry, National Taiwan University, Taipei City 10617, Taiwan
| |
Collapse
|
189
|
Bagrova O, Lapshina K, Sidorova A, Shpigun D, Lutsenko A, Belova E. Secondary structure analysis of proteins within the same topology group. Biochem Biophys Res Commun 2024; 734:150613. [PMID: 39222577 DOI: 10.1016/j.bbrc.2024.150613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The native conformation of a protein plays a decisive role in ensuring its functionality. It is established that the spatial structure of proteins may exhibit a greater degree of conservation than the corresponding amino acid sequences. This study aims to clarify structural distinctions between homologous and non-homologous proteins with identical topology. The analysis focuses on secondary structures with special emphasis on their fraction, distribution along the polypeptide chain, and chirality. Three different groups of proteins with identical topology were considered according to the CATH database: a homologous group of Globins, a group of Phycocyanins, which is often considered as a potential relative of globins, and a diverse assembly of other globin-like proteins. Some structural patterns in the distribution of secondary structure have been identified within Globins. A similar profile was observed in Phycocyanins, in contrast to the third group. In addition, a distinguishable structural motif, including structures such as 310-helix and irregular structure, has been found in both Globins and Phycocyanins, which can be proposed as an evolutionary imprint.
Collapse
Affiliation(s)
- Olga Bagrova
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Ksenia Lapshina
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alla Sidorova
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Denis Shpigun
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Aleksey Lutsenko
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Ekaterina Belova
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
190
|
Baothman O, Ali EMM, Alguridi H, Hosawi S, Konozy EHE, Abu Zeid IM, Ahmad A, Altayb HN. Impact of camel milk lactoferrin peptides against breast cancer cells: in silico and in vitro study. Front Pharmacol 2024; 15:1425504. [PMID: 39629082 PMCID: PMC11612555 DOI: 10.3389/fphar.2024.1425504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/23/2024] [Indexed: 12/06/2024] Open
Abstract
Background and Aims Breast cancer remains a significant global health concern, necessitating the exploration of novel therapeutic strategies. Despite advancements in cancer therapeutics, effective treatments with minimal side effects remain elusive. Natural sources, such as camel milk, harbor bioactive compounds such as lactoferrin peptides, which hold promise as anticancer agents. This study investigated the potential of camel milk-derived lactoferrin peptides against breast cancer cells through a combined in silico and in vitro approach. By integrating computational modeling with experimental assays, we aimed to elucidate the anticancer mechanisms of these peptides and provide insights for their optimization as anticancer therapeutics. Methods In silico analysis involving pepetid design, and validation, then molecular docking and molecular dynamics (MD) simulations was used to explore peptide-protein interactions and stability. Peptides were synthesized and tested for anticancer activity using MTT assays on MCF-7 cells, with HDFa normal cells used as controls. Results Results of this study showed that camel milk-derived lactoferrin peptides, particularly PEP66, exhibited strong anticancer activity against MCF-7 breast cancer cells, with the lowest IC50 value (52.82 μg/mL) compared to other peptides. In silico molecular docking and dynamics simulations revealed that PEP66 formed stable interactions with key residues in the HER2 catalytic site, indicating its potential as an effective anticancer agent. The selectivity index (SI) of PEP66 (3.19) also suggested lower toxicity to normal cells compared to cancer cells, reinforcing its therapeutic potential. Hydrogen bonding analysis highlighted key residues involved in stabilizing peptide-protein complexes, while molecular dynamics simulations demonstrated the stability of these interactions over time. Notably, PEP66 exhibited the highest stability and formed significant interactions with essential residues in the HER2 catalytic site, suggesting its potential as an effective anticancer agent. Conclusion Camel milk-derived lactoferrin peptides show promise as anticancer agents against breast cancer cells. The multidisciplinary approach employed in this study provides valuable insights into the mechanisms underlying their activity, paving the way for rational design strategies to enhance their efficacy. Further experimental validation is warranted to validate the anticancer potential of these peptides and advance their development as novel therapeutic agents for breast cancer treatment.
Collapse
Affiliation(s)
- Othman Baothman
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ehab M. M. Ali
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Division of Biochemistry, Chemistry Department, Faculty of Science Tanta University, Tanta, Egypt
| | - Hassan Alguridi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Emadeldin Hassan E. Konozy
- Laboratory of Proteomics and Glycobiology, Biotechnology Department, Africa City of Technology, Khartoum, Sudan
- Biomedical and Clinical Research Centre (BCRC), College of Health and Allied Sciences (CoHAS), University of Cape Coast, Cape Coast, Ghana
| | - Isam M. Abu Zeid
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abrar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hisham N. Altayb
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
191
|
Stephanie F, Tambunan USF, Kuczera K, Siahaan TJ. Structure of a Cyclic Peptide as an Inhibitor of Mycobacterium tuberculosis Transcription: NMR and Molecular Dynamics Simulations. Pharmaceuticals (Basel) 2024; 17:1545. [PMID: 39598454 PMCID: PMC11597662 DOI: 10.3390/ph17111545] [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: 12/11/2023] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND AND OBJECTIVES A novel antitubercular cyclic peptide, Cyclo(1,6)-Ac-CLYHFC-NH2, was designed to bind at the rifampicin (RIF) binding site on the RNA polymerase (RNAP) of Mycobacterium tuberculosis (MTB). This peptide inhibits RNA elongation in the MTB transcription initiation assay in the nanomolar range, which can halt the MTB transcription initiation complex, similar to RIF. Therefore, determining the solution conformation of this peptide is useful in improving the peptide's binding affinity to the RNAP. METHODS Here, the solution structure of Cyclo(1,6)-Ac-CLYHFC-NH2 was determined by two-dimensional (2D) NMR experiments and NMR-restrained molecular dynamic (MD) simulations. RESULTS All protons of Cyclo(1,6)-Ac-CLYHFC-NH2 were assigned using TOCSY and NOE NMR spectroscopy. The NOE cross-peak intensities were used to calculate interproton distances within the peptide. The JNH-HCα coupling constants were used to determine the possible Phi angles within the peptide. The interproton distances and calculated Phi angles from NMR were used in NMR-restrained MD simulations. The NOE spectra showed NH-to-NH cross-peaks at Leu2-to-Tyr3 and Tyr3-to-His4, indicating a βI-turn formation at the Cys1-Leu2-Tyr3-His4 sequence. CONCLUSIONS The NMR-restrained MD simulations showed several low-energy conformations that were congruent with the NMR data. Finally, the conformation of this peptide will be used to design derivatives that can better inhibit RNAP activity.
Collapse
Affiliation(s)
- Filia Stephanie
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Department of Chemistry, University of Indonesia, Depok 16424, Indonesia;
| | | | - Krzysztof Kuczera
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA;
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
192
|
Almohanna N, Koam ANA, Alhulwah K, Ahmad A, Azeem M. Characterization of entropy measures with connection number based indices of boric acid hydrogen-bonded 2D lattice sheets. Sci Rep 2024; 14:27807. [PMID: 39537831 PMCID: PMC11561266 DOI: 10.1038/s41598-024-79168-7] [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: 07/27/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
The cut method is a computational approach utilized to predict the fundamental activities of physicochemical properties of chemical networks, also called topological indices. The connection number is a new idea, that gives interesting and good results of the topological indices (TIs) and entropy measures (EMs) for structural representation of chemical compounds and networks. The physical density of chemical networks is characterized by these indices. In this paper, we determined the computational results for indices based on connection numbers for a two-dimensional lattice sheet of hydrogen-bonded boric acid. Boric acid, an inorganic compound, is not very harmful when applied to the skin or consumed. Finally, graphical and numerical comparisons of topological numbers including the number of borate hydrogen-bonded double lattice forms are also included in this study.
Collapse
Affiliation(s)
- Nasreen Almohanna
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia
| | - Ali N A Koam
- Department of Mathematics, College of Science, Jazan University, 114, 45142, Jazan, Kingdom of Saudi Arabia
| | - Khawlah Alhulwah
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia.
| | - Ali Ahmad
- Department of Computer Science, College of Engineering and Computer Science, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Muhammad Azeem
- Department of Mathematics, Riphah International University, Lahore, 54000, Pakistan
| |
Collapse
|
193
|
Adewale OO, Wińska P, Piasek A, Cieśla J. The Potential of Plant Polysaccharides and Chemotherapeutic Drug Combinations in the Suppression of Breast Cancer. Int J Mol Sci 2024; 25:12202. [PMID: 39596268 PMCID: PMC11594611 DOI: 10.3390/ijms252212202] [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: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Breast cancer is the most common cancer affecting women worldwide. The associated morbidity and mortality have been on the increase while available therapies for its treatment have not been totally effective. The most common treatment, chemotherapy, sometimes has dangerous side effects because of non-specific targeting, in addition to poor therapeutic indices, and high dose requirements. Consequently, agents with anticancer effects are being sought that can reduce the side effects induced by chemotherapy while increasing its cytotoxicity to cancer cells. This is possible using natural compounds that are safe and biologically active. There are many reports on plant polysaccharides due to their bioactive and anticancer properties. The use of plant polysaccharide together with a conventional cytotoxic drug may offer wide benefits in cancer therapy, producing synergistic effects, thereby reducing drug dose and, so, its associated side effects. In this review, we highlight an overview of the use of plant polysaccharides and chemotherapeutic drugs in breast cancer preclinical studies, including their mechanisms of anticancer activities. The findings emphasize the potential of plant polysaccharides to improve chemotherapeutic outcomes in breast cancer, paving the way for more effective and safer treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Joanna Cieśla
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.O.A.); (P.W.); (A.P.)
| |
Collapse
|
194
|
Nowitzke J, Bista S, Raman S, Dahal N, Stirnemann G, Popa I. Mechanical Unfolding of Network Nodes Drives the Stress Response of Protein-Based Materials. ACS NANO 2024; 18:31031-31043. [PMID: 39487800 DOI: 10.1021/acsnano.4c07352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
Biomaterials synthesized from cross-linked folded proteins have untapped potential for biocompatible, resilient, and responsive implementations, but face challenges due to costly molecular refinement and limited understanding of their mechanical response. Under a stress vector, these materials combine the gel-like response of cross-linked networks with the mechanical unfolding and extension of proteins from well-defined 3D structures to unstructured polypeptides. Yet the nanoscale dynamics governing their viscoelastic response remains poorly understood. This lack of understanding is further exacerbated by the fact that the mechanical stability of protein domains depends not only on their structure, but also on the direction of the force vector. To this end, here we propose a coarse-grained network model based on the physical characteristics of polyproteins and combine it with the mechanical unfolding response of protein domains, obtained from single molecule measurements and steered molecular dynamics simulations, to explain the macroscopic response of protein-based materials to a stress vector. We find that domains are about 10-fold more stable when force is applied along their end-to-end coordinate than along the other tethering geometries that are possible inside the biomaterial. As such, the macroscopic response of protein-based materials is mainly driven by the unfolding of the node-domains and rearrangement of these nodes inside the material. The predictions from our models are then confirmed experimentally using force-clamp rheometry. This model is a critical step toward developing protein-based materials with predictable response and that can enable applications for shape memory and energy storage and dissipation.
Collapse
Affiliation(s)
- Joel Nowitzke
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Sanam Bista
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Sadia Raman
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Narayan Dahal
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Guillaume Stirnemann
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne University, CNRS, Paris 75005, France
| | - Ionel Popa
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
195
|
He S, Taher N, Simard A, Hvorecny K, Ragusa M, Bahl C, Hickman A, Dyda F, Madden D. Molecular basis for the transcriptional regulation of an epoxide-based virulence circuit in Pseudomonas aeruginosa. Nucleic Acids Res 2024; 52:12727-12747. [PMID: 39413156 PMCID: PMC11648964 DOI: 10.1093/nar/gkae889] [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: 01/15/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa infects the airways of people with cystic fibrosis (CF) and produces a virulence factor Cif that is associated with worse outcomes. Cif is an epoxide hydrolase that reduces cell-surface abundance of the cystic fibrosis transmembrane conductance regulator (CFTR) and sabotages pro-resolving signals. Its expression is regulated by a divergently transcribed TetR family transcriptional repressor. CifR represents the first reported epoxide-sensing bacterial transcriptional regulator, but neither its interaction with cognate operator sequences nor the mechanism of activation has been investigated. Using biochemical and structural approaches, we uncovered the molecular mechanisms controlling this complex virulence operon. We present here the first molecular structures of CifR alone and in complex with operator DNA, resolved in a single crystal lattice. Significant conformational changes between these two structures suggest how CifR regulates the expression of the virulence gene cif. Interactions between the N-terminal extension of CifR with the DNA minor groove of the operator play a significant role in the operator recognition of CifR. We also determined that cysteine residue Cys107 is critical for epoxide sensing and DNA release. These results offer new insights into the stereochemical regulation of an epoxide-based virulence circuit in a critically important clinical pathogen.
Collapse
Affiliation(s)
- Susu He
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Noor M Taher
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Adam R Simard
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Kelli L Hvorecny
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Michael J Ragusa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
- Department of Chemistry, Dartmouth, Hanover, NH 03755, USA
| | - Christopher D Bahl
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Alison B Hickman
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fred Dyda
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dean R Madden
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
- Department of Chemistry, Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
196
|
Göring J, Schwarz C, Unger E, Quaas R, Hilger I. The Long-Term Impact of Polysaccharide-Coated Iron Oxide Nanoparticles on Inflammatory-Stressed Mice. J Xenobiot 2024; 14:1711-1728. [PMID: 39584956 PMCID: PMC11587046 DOI: 10.3390/jox14040091] [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: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Since iron oxide nanoparticles (IONPs) are expected to be important tools in medical care, patients with inflammatory diseases will be increasingly exposed to IONPs in the future. Here, we assessed the short- and long-term impact of polysaccharide (PS)-coated IONPs on mice with persistent systemic inflammation. To this end, PS-IONPs were synthetized by a core-shell method. Mice were regularly injected with sterile zymosan. PS-IONPs were administered intravenously. At specific nanoparticle injection post-observation times, the organ iron concentration was determined via atomic absorption spectrometry, the expression of NF-κB-related proteins using SDS-PAGE and immunoblotting, as well as body weight and haemograms. Finally, the mediator secretion in blood plasma was analysed using multiplexed ELISA. Our data show that PS-IONPs induce short-term changes of iron levels in distinct organs and of NF-κB p65 and p50, p100, COX-2s, and Bcl-2 protein expression in the liver of inflammatory stressed mice. In the long term, there was an attenuated expression of several NF-κB-related proteins and attenuated features of inflammatory-based anaemia in blood. PS-IONPs weakly influenced the blood cytokine levels. PS-IONPs are biocompatible, but given their short-term pro-inflammatory impact, they should prospectively be applied with caution in patients with inflammatory diseases of the liver.
Collapse
Affiliation(s)
- Julia Göring
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (J.G.)
| | - Claudia Schwarz
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (J.G.)
| | - Eric Unger
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (J.G.)
| | - Rainer Quaas
- Chemicell GmbH, Eresburgstrasse 22-23, D-12103 Berlin, Germany;
| | - Ingrid Hilger
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (J.G.)
| |
Collapse
|
197
|
Wang R, Tursun M, Shan W. Complete Mitogenomes of Xinjiang Hares and Their Selective Pressure Considerations. Int J Mol Sci 2024; 25:11925. [PMID: 39595994 PMCID: PMC11593953 DOI: 10.3390/ijms252211925] [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: 10/05/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Comparative analysis based on the mitogenomes of hares in Xinjiang, China, is limited. In this study, the complete mitochondrial genomes of seven hare samples including four hare species and their hybrids from different environments were sequenced, assembled, and annotated. Subsequently, we performed base content and bias analysis, tRNA analysis, phylogenetic analysis, and amino acid sequence analysis of the annotated genes to understand their characteristics and phylogenetic relationship. Their mitogenomes are circular molecules (from 16,691 to 17,598 bp) containing 13 protein-coding genes, two rRNA genes, 22 tRNA genes, and a control region, which are similar with other Lepus spp. worldwide. The relative synonymous codon usage analysis revealed that the adaptation of Lepus yarkandensis to its unique arid and hot environment might be associated with synthesizing amino acids like alanine, leucine, serine, arginine, and isoleucine and the terminator caused by the different usage of codons. Further, we utilized the MEME model and identified two positive selection genes (ND4, ND5) in Lepus tibetanus pamirensis and one (ND5) in L. yarkandensis that might be important to their adaptation to the plateau and dry and hot basin environments, respectively. Meanwhile, Lepus tolai lehmanni and Lepus timidus may have evolved different adaptive mechanisms for the same cold environment. This study explored the evolutionary dynamics of Xinjiang hares' mitochondrial genomes, providing significant support for future research into their adaptation mechanisms in extreme environments.
Collapse
Affiliation(s)
| | | | - Wenjuan Shan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (R.W.); (M.T.)
| |
Collapse
|
198
|
Bist PS, Tayara H, Chong KT. Generative AI in the Advancement of Viral Therapeutics for Predicting and Targeting Immune-Evasive SARS-CoV-2 Mutations. IEEE J Biomed Health Inform 2024; 28:6974-6982. [PMID: 39042543 DOI: 10.1109/jbhi.2024.3432649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The emergence of immune-evasive mutations in the SARS-CoV-2 spike protein is consistently challenging existing vaccines and therapies, making precise prediction of their escape potential a critical imperative. Artificial Intelligence(AI) holds great promise for deciphering the intricate language of protein. Here, we employed a Generative Adversarial Network to decipher the hidden escape pathways within the spike protein by generating spikes that closely resemble natural ones. Through comprehensive analysis, we demonstrated that generated sequences capture natural escape characteristics. Moreover, incorporating these sequences into an AI-based escape prediction model significantly enhanced its performance, achieving a 7% increase in detecting natural escape mutations on the experimentally validated Greaney dataset. Similar improvements were observed on other datasets, demonstrating the model's generalizability. Precisely predicting immune-evasive spikes not only enables the design of strategically targeted therapies but also has the potential to expedite future viral therapeutics. This breakthrough carries profound implications for shaping a more resilient future against viral threats.
Collapse
|
199
|
Arshad F, Sarfraz A, Rubab A, Shehroz M, Moura AA, Sheheryar S, Ullah R, Shahat AA, Ibrahim MA, Nishan U, Shah M. Rational design of novel peptide-based vaccine against the emerging OZ virus. Hum Immunol 2024; 85:111162. [PMID: 39447523 DOI: 10.1016/j.humimm.2024.111162] [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: 08/27/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Oz virus (OZV) belongs to the Orthomyxoviridae family which includes viruses with a negative-sense, single-stranded, and segmented RNA genome. OZV is a zoonotic pathogen, particularly since the virus can cause deadly illness when injected intracerebrally into nursing mice. OZV is an emerging pathogen with the potential to spark a pandemic as there is no preventive and licensed treatment against this virus. The goal of this study was to develop a novel multi-epitope vaccination against OZV proteins utilizing immunoinformatics and immunological simulation analysis. This work evaluated immunological epitopes (B cells, MHC-I, and MHC-II) to identify highly antigenic OZV target proteins. Shortlisted epitopes were joined together by using appropriate linkers and adjuvants to design multi-epitope vaccine constructs (MEVC). The vaccine models were designed, improved, validated, and the globular regions and post-translational modifications (PTMs) were also evaluated in the vaccine's structure. Molecular docking analysis with the Toll-like receptor (TLR4) showed strong interactions and appropriate binding energies. Molecular dynamics (MD) simulation confirmed stable interactions between the vaccines and TLR4. Bioinformatics tools helped optimize codons, resulting in successful cloning into appropriate host vectors. This study showed that the developed vaccines are stable and non-allergenic in the human body and successfully stimulated immunological responses against OZV. Finally, a mechanism of action for the designed vaccine construct was also proposed. Further experimental validations of the designed vaccine construct will pave the way to create a potentially effective vaccine against this emerging pathogen.
Collapse
Affiliation(s)
- Fizza Arshad
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Asifa Sarfraz
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Aleeza Rubab
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Muhammad Shehroz
- Department of Bioinformatics, Kohsar University Murree, Murree 47150, Pakistan
| | - Arlindo A Moura
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil
| | - Sheheryar Sheheryar
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaaty A Shahat
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Ibrahim
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Umar Nishan
- Department of Chemistry, Kohat University of Science & Technology, Kohat, Pakistan.
| | - Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan; Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil.
| |
Collapse
|
200
|
Wohl S, Gilron Y, Zheng W. Structural and Functional Relevance of Charge Based Transient Interactions inside Intrinsically Disordered Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621161. [PMID: 39554085 PMCID: PMC11565980 DOI: 10.1101/2024.10.30.621161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Intrinsically disordered proteins (IDPs) perform a wide range of biological functions without adopting stable, well-defined, three-dimensional structures. Instead, IDPs exist as dynamic ensembles of flexible conformations, traditionally thought to be governed by weak, nonspecific interactions, which are well described by homopolymer theory. However, recent research highlights the presence of transient, specific interactions in several IDPs, suggesting that factors beyond overall size influence their conformational behavior. In this study, we investigate how the spatial arrangement of charged amino acids within IDP sequences shapes the prevalence of transient, specific interactions. Through a series of model peptides, we establish a quantitative empirical relationship between the fraction of transient interactions and a novel sequence metric, termed effective charged patch length, which characterizes the ability of charged patches to drive these interactions. By examining IDP ensembles with varying levels of transient interactions, we further explore their heteropolymeric structural behavior in phase-separated condensates, where we observe the formation of a condensate-spanning network structure. Additionally, we perform a proteome-wide scan for charge-based transient interactions within disordered regions of the human proteome, revealing that approximately 10% of these regions exhibit such charge-driven transient interactions, leading to heteropolymeric behaviors in their conformational ensembles. Finally, we examine how these charge-based transient interactions correlate with molecular functions, identifying specific biological roles in which these interactions are enriched.
Collapse
Affiliation(s)
- Samuel Wohl
- Department of Physics, Arizona State University, Tempe, AZ 85287, USA
| | - Yishai Gilron
- College of Integrative Sciences and Arts, Arizona State University, Mesa, AZ 85212, USA
| | - Wenwei Zheng
- College of Integrative Sciences and Arts, Arizona State University, Mesa, AZ 85212, USA
| |
Collapse
|