1
|
Yang W, Feng X, Chen H, Liman GLS, Santangelo TJ, Zhang C, Zeng Z. Cyclization of archaeal membrane lipids impacts membrane protein activity and archaellum formation. Proc Natl Acad Sci U S A 2025; 122:e2423648122. [PMID: 40354536 DOI: 10.1073/pnas.2423648122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Enhancement of the cyclization of membrane lipids GDGTs (glycerol dialkyl glycerol tetraethers) is a critical strategy for archaea to adapt to various environmental stresses. However, the physiological function of membrane lipid cyclization remains unclear. Here, we reported that the GDGT ring synthases mutant, deficient in GDGT cyclization, inhibited archaellum formation and reduced cell motility in thermoacidophilic crenarchaeon Sulfolobus acidocaldarius. This inhibition was caused by decreased transcription of the archaellum operon, likely due to cleavage of the C-terminal domains in transmembrane proteins ArnRs, the transcription factors that regulate archaellum operon expression. The transcriptomic and proteomic analysis showed deficiency of GDGT cyclization broadly impacted the expression of membrane associate proteins, including respiratory chain proteins, and decreased cellular ATP concentration. Moreover, phylogenetic analysis demonstrated that the correlation between GDGT cyclization and archaellum formation is widespread among (hyper)thermophilic archaea, and this was further verified in the euryarchaeon Thermococcus kodakarensis. Our findings suggested that archaea modify their membrane lipids to profoundly alter cellular appendages and cell physiology to adapt to environmental fluctuations.
Collapse
Affiliation(s)
- Wei Yang
- Department of Ocean Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Feng
- Department of Ocean Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huahui Chen
- Department of Ocean Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Thomas J Santangelo
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Changyi Zhang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Zhirui Zeng
- Department of Ocean Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
2
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025; 125:4528-4568. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [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: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
3
|
Tripathi A, Chhabra A, Rizvi S, Tyagi RK. Selective steroid receptor modulators, degraders and PROTACs: Therapeutic strategies in management of endocrine-related cancers. Mol Cell Endocrinol 2025; 605:112569. [PMID: 40339978 DOI: 10.1016/j.mce.2025.112569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/23/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Endocrine-related disorders are highly prevalent globally, affecting millions of people. Such diseases are multifactorial in origin and are influenced by the complex interplay of genetics, lifestyle, and environmental factors. Recurring disruptions in the endocrine homeostasis can lead to a cascade of endocrine-related cancers. It is well known that nuclear receptors (NRs), particularly estrogen receptor and androgen receptor malfunctioning promote the oncogenesis of breast cancer and prostate cancer, respectively. However, existing therapeutics against these diseases, including aromatase inhibitors, (anti-) hormonal therapy, etc., often yield limited success, prompting to explore alternative methods of disease management. Additionally, drug resistance is prominent in cancer patients undergoing multidrug therapy. Currently, novel drug design strategies targeting NRs are being implemented for the discovery of a new generation of small molecule modulators, including selective NR modulators (SNuRMs) and degraders (SNuRDs). Moreover, proteolysis-targeting chimeras (PROTACs) as NR degraders, are also being developed primarily to overcome drug resistance, enhance protein selectivity, and mitigate off-target toxicity. This review highlights recent advancements in SNuRMs and SNuRDs for managing NRs-associated endocrine/metabolic disorders. Furthermore, we discuss the therapeutic potential of PROTAC degraders as a stand-alone strategy for receptor-mediated disease intervention, offering new avenues for precision medicine.
Collapse
Affiliation(s)
- Anjali Tripathi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Ayushi Chhabra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Sheeba Rizvi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Rakesh K Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India.
| |
Collapse
|
4
|
Nguyen KA, Falson P, Boumendjel A. Assembly of Detergents with Highly Branched Dicarboxylate Clamps for Membrane Protein Studies. ChemistryOpen 2025:e2500122. [PMID: 40326160 DOI: 10.1002/open.202500122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/03/2025] [Indexed: 05/07/2025] Open
Abstract
The structural knowledge of membrane proteins (MPs) is crucial for the structure-based drug design. The stabilization of MPs during extraction processes is essential for structural and functional maintenance. In this regard, detergents are used to achieve extractions of MPs in their functional form. Based on previous work showing the importance of adjacent dicarboxylate groups for the detergency properties, the synthesis of a new generation of detergents bearing more dicarboxylate groups is reported. The molecular structure of the new generation is characterized by the presence of four chemical entities: a DOTA or NOTA scaffold, three or four pairs of carboxylates, a fatty acid, and PEG chains. The preliminary biochemical evaluation reveals promising features of this novel generation of surfactants for the aqueous extraction of MPs.
Collapse
Affiliation(s)
- Kim-Anh Nguyen
- Laboratoire Radiopharmaceutiques Biocliniques, LRB, Faculté de Médecine, INSERM, Domaine de la Merci, 38700, La Tronche, France
| | - Pierre Falson
- Drug Resistance & Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory (CNRS UMR 5086), IBCP, University of Lyon, 7 Passage du Vercors, 69367, Lyon, France
| | - Ahcène Boumendjel
- Laboratoire Radiopharmaceutiques Biocliniques, LRB, Faculté de Médecine, INSERM, Domaine de la Merci, 38700, La Tronche, France
| |
Collapse
|
5
|
Sun J, Saimi M, Rempel D, Cao Q, Chai M, Li W, Gross ML. In-Cell Fast Photochemical Oxidation Interrogates the Native Structure of Integral Membrane Proteins. Angew Chem Int Ed Engl 2025; 64:e202424779. [PMID: 40033852 PMCID: PMC12052488 DOI: 10.1002/anie.202424779] [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: 12/17/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
Integral membrane proteins (IMPs) are pivotal for cellular functions but challenging to investigate. Here, IC-FPOMP (in-cell fast photochemical oxidation of MPs) is introduced, a method enabling in situ footprinting of IMPs within live cells. IC-FPOMP generates reactive oxygen radicals from various precursors (TiO2 nanoparticles or H2O2) near the membrane. Leveraging a laser and a 96-well plate platform, high-throughput and rapid footprinting of IMPs are achieved. IC-FPOMP of two human IMPs (human glucose transporter-hGLUT1 and human gamma-glutamyl carboxylase-hGGCX) are successful, providing footprinting of both the transmembrane and extramembrane regions. Comparative analysis of hGLUT1 in liposomes versus cells shows that the membrane may impact the transporter's conformation differently. In-cell drug screening targeting hGLUT1 reveals drug-binding behavior in vivo. In summary, IC-FPOMP offers insights into IMP structure-function relationships in cells and facilitates drug discovery.
Collapse
Affiliation(s)
- Jie Sun
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, Box 1134, St. Louis, MO, 63130, USA
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, 1311 Cumberland Avenue, Knoxville, TN, 37996-1937, USA
| | - Mierxiati Saimi
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave, Box 8231, St. Louis, MO, 63110, USA
| | - Don Rempel
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, Box 1134, St. Louis, MO, 63130, USA
| | - Qing Cao
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave, Box 8231, St. Louis, MO, 63110, USA
| | - Mengqi Chai
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, Box 1134, St. Louis, MO, 63130, USA
| | - Weikai Li
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave, Box 8231, St. Louis, MO, 63110, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, Box 1134, St. Louis, MO, 63130, USA
| |
Collapse
|
6
|
Takács G, Balogh GT, Kiss R. A data-driven journey using results from target-based drug discovery for target deconvolution in phenotypic screening. RSC Med Chem 2025:d4md01051e. [PMID: 40352671 PMCID: PMC12062751 DOI: 10.1039/d4md01051e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/12/2025] [Indexed: 05/14/2025] Open
Abstract
In drug discovery, various approaches exist to find compounds that alter the different states in living organisms. There are two fundamental discovery strategies regarding the mechanism of action: target-based and phenotypic methods. Both have strengths and weaknesses in assay development, target selection, target validation and structure optimization. While phenotypic screening can identify chemical starting points with the desired phenotype, it is typically difficult to carry out efficient, structure-based optimization without confirming the mechanism of action of such hits. It is therefore critical to uncover the targets behind the phenotype. Target deconvolution is typically carried out by a set of highly selective compounds, where each ligand is associated with a particular target. Hits of such a high-selectivity set can provide valuable information on the phenotype's underlying targets and may also enable novel target-based therapeutic strategies. Consequently, there is a continuously high demand for novel highly-selective tool compounds for target deconvolution. In this work, the ChEMBL database, comprising over 20 million bioactivity data, was mined to identify the most selective novel ligands for a diverse set of targets. A novel method for the automated selection of such high-selectivity ligands is presented. Using these high-selectivity compounds in phenotypic screening campaigns can provide a valuable preliminary direction during target deconvolution. 87 representative compounds were purchased and screened against 60 cancer cell lines. Several compounds were found to possess selective inhibition of cell growth of a few distinct cell lines. The phenotypic assay results, along with the nanomolar activities of individual proteins obtained from the ChEMBL database suggest some novel mechanisms of action for anti-cancer drug discovery.
Collapse
Affiliation(s)
- Gergely Takács
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics Műegyetem Rakpart 3 Budapest 1111 Hungary
- Mcule.com Kft Bartók Béla út 105-113 Budapest 1115 Hungary
| | - György T Balogh
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics Műegyetem Rakpart 3 Budapest 1111 Hungary
- University Pharmacy Department of Pharmacy Administration, Semmelweis University 7-9 Hőgyes Street 1092 Budapest Hungary
| | - Róbert Kiss
- Mcule.com Kft Bartók Béla út 105-113 Budapest 1115 Hungary
| |
Collapse
|
7
|
Sellami A, Montes M, Lagarde N. Combining Molecular Docking and Pharmacophore Models Predicts Ligand Binding of Endocrine-Disrupting Chemicals to Nuclear Receptors. Chem Res Toxicol 2025; 38:672-685. [PMID: 40178351 DOI: 10.1021/acs.chemrestox.4c00455] [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: 04/05/2025]
Abstract
Nuclear receptors form a family of proteins capable of accommodating a wide variety of small molecules in their ligand binding domain, ranging from therapeutic compounds to endocrine-disrupting chemicals. The rapid identification of these compounds, especially within the latter category, is of paramount importance. Using data extracted from the CompTox Dashboard, an Environmental Protection Agency initiative, we assessed the effectiveness of a combination of molecular docking and pharmacophore models in identifying ligands binding to six nuclear receptors: androgen receptor, estrogen receptor alpha, estrogen receptor beta, glucocorticoid receptor, peroxisome proliferator-activated receptor gamma, and thyroid hormone receptor alpha. For each nuclear receptor, we selected a specifically designed and optimized in silico protocol that, in conjunction with experimental assays, can prioritize compounds for further evaluation to detect any potential toxicological concerns.
Collapse
Affiliation(s)
- Asma Sellami
- Laboratoire GBCM, EA 7528, Conservatoire National des Arts et Métiers, 2 rue Conté, Paris 75003, France
| | - Matthieu Montes
- Laboratoire GBCM, EA 7528, Conservatoire National des Arts et Métiers, 2 rue Conté, Paris 75003, France
- Institut Universitaire de France, Paris 75003, France
| | - Nathalie Lagarde
- Laboratoire GBCM, EA 7528, Conservatoire National des Arts et Métiers, 2 rue Conté, Paris 75003, France
| |
Collapse
|
8
|
Youn T, Kim G, Hariharan P, Li X, Ahmed W, Byrne B, Liu X, Guan L, Chae PS. Improved Pendant-Bearing Glucose-Neopentyl Glycols for Membrane Protein Stability. Bioconjug Chem 2025; 36:707-717. [PMID: 40105011 DOI: 10.1021/acs.bioconjchem.4c00556] [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: 03/20/2025]
Abstract
Membrane proteins are biologically and pharmaceutically significant, and determining their 3D structures requires a membrane-mimetic system to maintain protein stability. Detergent micelles are widely used as membrane mimetics; however, their dynamic structures often lead to the denaturation and aggregation of encapsulated membrane proteins. To address the limitations of classical detergents in stabilizing membrane proteins, we previously reported a class of glucose-neopentyl glycols (GNGs) and their pendant-bearing versions (P-GNGs), several of which proved more effective than DDM in stabilizing membrane proteins. In this study, we synthesized additional GNG derivatives by varying the lengths of the pendant (P-GNGs), and by introducing hemifluorinated pendants to the GNG scaffold (fluorinated pendant-bearing GNGs or FP-GNGs). The synthetic flexibility of the GNG chemical architecture allowed us to create a diverse range of derivatives, essential for the effective optimization of detergent properties. When tested with two model membrane proteins (a transporter and a G-protein coupled receptor (GPCR)), most of the new (F)P-GNGs demonstrated superior stabilization of these membrane proteins compared to DDM, the original GNG (OGNG)), and a previously developed P-GNG (i.e., GNG-3,14). Notably, several P-GNGs synthesized in this study were as effective as or even better than lauryl maltose neopentyl glycol (LMNG) in stabilizing a human GPCR, beta2 adrenergic receptor (β2AR). Enhanced protein stability was particularly observed for the P-GNGs with a butyl (C4) or pentyl (C5) pendant, indicating that these pendant sizes are optimal for membrane protein stability. The volumes of these pendants appear to minimize the empty spaces in the micelle interiors, thereby enhancing detergent-detergent interactions in micelles complexed with the membrane proteins. Additionally, we identified one FP-GNG that was more efficient at extracting the transporter and more effective at stabilizing the GPCR than DDM. Thus, the current study demonstrates that both chain length and number of fluorine atoms in the pendants of the P-GNGs were crucial determinants for membrane protein stability. We not only developed a few (F)P-GNGs that are significantly more effective than maltoside detergents (LMNG/DDM) for protein extraction and stability but we also provided an effective strategy for detergent design through the utilization of partially fluorinated pendants of varying length.
Collapse
Affiliation(s)
- Taeyeol Youn
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| | - Ganghee Kim
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| | - Parameswaran Hariharan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Xianglan Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Waqar Ahmed
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Xiangyu Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Pil Seok Chae
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| |
Collapse
|
9
|
Guo J, Hou Q, Tan Y, Fu R, Huang X, Cao C. Membrane Proteins in Nanodiscs: Methods and Applications. ChemMedChem 2025; 20:e202400775. [PMID: 39825697 DOI: 10.1002/cmdc.202400775] [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: 09/29/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/20/2025]
Abstract
Membrane proteins, a principal class of drug targets, play indispensable roles in various biological processes and are closely associated with essential life functions. Their study, however, is complicated by their low solubility in aqueous environments and distinctive structural characteristics, necessitating a suitable native-like environment for molecular analysis. Nanodisc technology has revolutionized this field, providing biochemists with a powerful tool to stabilize membrane proteins and significantly enhance their research possibilities. This review outlines the substantial advancements in nanodisc methodologies and applications from 2018 to 2024. We cover the development of various nanodisc models, as well as structural and functional studies of membrane proteins that utilize nanodiscs, highlighting their medical applications.
Collapse
Affiliation(s)
- Jiaxu Guo
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science, Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Qinghan Hou
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, 91125, US
| | - Yulin Tan
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science, Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Ruoheng Fu
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science, Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xuanwei Huang
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science, Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chan Cao
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science, Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
10
|
Sadaf A, Yun HS, Lee H, Stanfield S, Lan B, Salomon K, Woubshete M, Kim S, Ehsan M, Bae H, Byrne B, Loland CJ, Liu X, Guan L, Im W, Chae PS. Multiple Pendants-Bearing Triglucosides for Membrane Protein Studies: Effects of Pendant Length and Number on Micelle Interior Hydration and Protein Stability. Biomacromolecules 2025; 26:2565-2579. [PMID: 40087026 DOI: 10.1021/acs.biomac.5c00036] [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: 03/16/2025]
Abstract
Membrane proteins play central roles in cell physiology and are the targets of over 50% of FDA-approved drugs. In the present study, we prepared single alkyl-chained triglucosides decorated with multiple pendants, designated multiple pendant-bearing glucosides (MPGs), to enhance membrane protein stability. The new detergents feature two and four pendants of varying size at the hydrophilic-lipophilic interfaces, designated MPG-Ds and MPG-Ts, respectively. When tested with model membrane proteins, including the human adrenergic receptor (β2AR), the tetra-pendant-bearing MPGs (MPG-Ts) demonstrated superior performance compared to the dipendant analogs (MPG-Ds) and the gold standard DDM. All-atom molecular dynamics (MD) simulations results reveal that the four-pendant configuration of this detergent is remarkably effective in excluding water from the hydrophobic micelle interiors compared to the dipendant MPGs and DDM, an unprecedented feature of this new detergent. Our findings provide a novel strategy for designing water-resistant detergents, advancing the field of membrane protein research.
Collapse
Affiliation(s)
- Aiman Sadaf
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, Republic of Korea
| | - Hong Sik Yun
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, Republic of Korea
| | - Hajin Lee
- MolCube, Inc., Seocho-gu, Seoul 06640, Republic of Korea
| | - Samantha Stanfield
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Baoliang Lan
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Kristine Salomon
- Department of Neuroscience, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Menebere Woubshete
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Seonghoon Kim
- MolCube, Inc., Seocho-gu, Seoul 06640, Republic of Korea
| | - Muhammad Ehsan
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, Republic of Korea
| | - Hyemi Bae
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, Republic of Korea
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Claus J Loland
- Department of Neuroscience, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Xiangyu Liu
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Wonpil Im
- MolCube, Inc., Seocho-gu, Seoul 06640, Republic of Korea
- Department of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Pil Seok Chae
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, Republic of Korea
| |
Collapse
|
11
|
Chien CT, Maduke M, Chiu W. Single-particle cryogenic electron microscopy structure determination for membrane proteins. Curr Opin Struct Biol 2025; 92:103047. [PMID: 40228430 DOI: 10.1016/j.sbi.2025.103047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/16/2025]
Abstract
Membrane proteins are crucial to many cellular functions but are notoriously difficult for structural studies due to their instability outside their natural environment and their amphipathic nature with dual hydrophobic and hydrophilic regions. Single-particle cryogenic electron microscopy (cryo-EM) has emerged as a transformative approach, providing near-atomic-resolution structures without the need for crystallization. This review discusses advancements in cryo-EM, emphasizing membrane sample preparation and data processing techniques. It explores innovations in capturing membrane protein structures within native environments, analyzing their dynamics, binding partner interactions, lipid associations, and responses to electrochemical gradients. These developments continue to enhance our understanding of these vital biomolecules, advancing the contributions of structural biology for basic and translational biomedicine.
Collapse
Affiliation(s)
- Chih-Ta Chien
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Merritt Maduke
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Wah Chiu
- Departments of Bioengineering, and of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Tang K, Tang Z, Niu M, Kuang Z, Xue W, Wang X, Liu X, Yu Y, Jeong S, Ma Y, Wu A, Kim BYS, Jiang W, Yang Z, Li C. Allosteric targeted drug delivery for enhanced blood-brain barrier penetration via mimicking transmembrane domain interactions. Nat Commun 2025; 16:3410. [PMID: 40210849 PMCID: PMC11986143 DOI: 10.1038/s41467-025-58746-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: 07/01/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Current strategies for active targeting in the brain are entirely based on the effective interaction of the ligand with the orthosteric sites of specific receptors on the blood-brain barrier (BBB), which is highly susceptible to various pathophysiological factors and limits the efficacy of drug delivery. Here, we propose an allosteric targeted drug delivery strategy that targets classical BBB transmembrane receptors by designing peptide ligands that specifically bind to their transmembrane domains. This strategy prevents competitive interference from endogenous ligands and antibodies by using the insulin receptor and integrin αv as model targets, respectively, and can effectively overcome pseudotargets or target loss caused by shedding or mutating the extracellular domain of target receptors. Moreover, these ligands can be spontaneously embedded in the phospholipid layer of lipid carriers using a plug-and-play approach without chemical modification, with excellent tunability and immunocompatibility. Overall, this allosteric targeted drug delivery strategy can be applied to multiple receptor targets and drug carriers and offers promising therapeutic benefits in brain diseases.
Collapse
Affiliation(s)
- Kaicheng Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Miaomiao Niu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zuyin Kuang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xinyu Wang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Yang Yu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Seongdong Jeong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Annette Wu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Wang L, Markus H, Chen D, Chen S, Zhang F, Gao S, Khunsriraksakul C, Chen F, Olsen N, Foulke G, Jiang B, Carrel L, Liu DJ. An atlas of single-cell eQTLs dissects autoimmune disease genes and identifies novel drug classes for treatment. CELL GENOMICS 2025; 5:100820. [PMID: 40154479 PMCID: PMC12008810 DOI: 10.1016/j.xgen.2025.100820] [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: 02/27/2024] [Revised: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Most variants identified from genome-wide association studies (GWASs) are non-coding and regulate gene expression. However, many risk loci fail to colocalize with expression quantitative trait loci (eQTLs), potentially due to limited GWAS and eQTL analysis power or cellular heterogeneity. Population-scale single-cell RNA-sequencing (scRNA-seq) datasets are emerging, enabling mapping of eQTLs in different cell types (sc-eQTLs). Compared to eQTL data from bulk tissues (bk-eQTLs), sc-eQTL datasets are smaller. We propose a joint model of bk-eQTLs as a weighted sum of sc-eQTLs (JOBS) from constituent cell types to improve power. Applying JOBS to One1K1K and eQTLGen data, we identify 586% more eQTLs, matching the power of 4× the sample sizes of OneK1K. Integrating sc-eQTLs with GWAS data creates an atlas for 14 immune-mediated disorders, colocalizing 29.9% or 32.2% more loci than using sc-eQTL or bk-eQTL alone. Extending JOBS, we develop a drug-repurposing pipeline and identify novel drugs validated by real-world data.
Collapse
Affiliation(s)
- Lida Wang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Havell Markus
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Dieyi Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Siyuan Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Fan Zhang
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Shuang Gao
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Chachrit Khunsriraksakul
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Fang Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nancy Olsen
- Department of Medicine, Penn State University, College of Medicine, Hershey, PA 17033, USA
| | - Galen Foulke
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Dermatology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Bibo Jiang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Laura Carrel
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Dajiang J Liu
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
14
|
Dai Y, Yang M, Luo W, Qiu Y, Zhou F, Zheng X, Zhao F, Yao X, Zhao S, Tao H. "Living Detergents": an in Situ Detergent Tailoring Strategy for Efficient Membrane Protein Stabilization and Analysis. Chemistry 2025:e202501128. [PMID: 40192258 DOI: 10.1002/chem.202501128] [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: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
Detergents are essential molecular tools for membrane protein (MP) research, yet traditional detergents with static properties often fail to address the diverse and evolving needs of MP studies. To this end, this study introduces "living detergents", an innovative class of detergents equipped with functional tags that enable bioorthogonal modifications with externally introduced structural elements. This approach allows for not only the parallel generation of new detergents, but also in situ tuning of MP samples within freshly formed detergents. The efficacy of this strategy was demonstrated through the rapid identification of optimal detergents for high-quality electron microscopy studies of A2AAR. Overall, this flexible and robust platform enables efficient tailoring of detergents, advancing the exploration of detergent structure-function relationships in MP research and opening pathways for more specialized solutions for diverse experimental demands.
Collapse
Affiliation(s)
- Yili Dai
- Institute of Pharmacy and Pharmacology, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Meifang Yang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weiling Luo
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Yanli Qiu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fang Zhou
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Xing Zheng
- Institute of Pharmacy and Pharmacology, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan, 410004, China
| | - Fei Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Xu Yao
- Institute of Pharmacy and Pharmacology, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Houchao Tao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
15
|
Breyer M, Lamer S, Schlosser A, Üçeyler N. Human sensory-like neuron surfaceome analysis. PLoS One 2025; 20:e0320056. [PMID: 40173182 PMCID: PMC11964241 DOI: 10.1371/journal.pone.0320056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/12/2025] [Indexed: 04/04/2025] Open
Abstract
Acral and triggerable pain is a hallmark of diseases involving small nerve fiber impairment, yet the underlying cellular mechanisms remain elusive. A key role is attributed to pain-related proteins located within the neuronal plasma membrane of nociceptive neurons. To explore this, we employed human induced pluripotent stem cell-derived sensory-like neurons and enriched their surface proteins by biotinylation. Samples from three independent cell differentiations were analyzed via liquid chromatography tandem mass spectrometry. Detected proteins were categorized by cellular location and function, followed by generating an interaction network for deregulated surface proteins. Gene expression of selected proteins was quantified using real-time PCR. A comparative analysis was performed between a patient with Fabry disease (FD) and a healthy control, which we used as model system. We successfully extracted surfaceome proteins from human sensory-like neurons, revealing deregulation of 48 surface proteins in FD-derived neurons. Among the candidates with potential involvement in pain pathophysiology were CACNA2D3, GPM6A, EGFR, and ABCA7. Despite the lack of gene expression differences in these candidates, the interaction network indicated compromised neuronal network integrity. Our approach successfully enabled the extraction and comprehensive analysis of the surfaceome from human sensory-like neurons, establishing a novel methodological framework for investigating human sensory-like neuron biology and cellular disease mechanisms.
Collapse
Affiliation(s)
- Maximilian Breyer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Stephanie Lamer
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
- Würzburg Fabry Center for Interdisciplinary Therapy (FAZIT), University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Hashimoto Y, Shil S, Tsuruta M, Kawauchi K, Miyoshi D. Three- and four-stranded nucleic acid structures and their ligands. RSC Chem Biol 2025; 6:466-491. [PMID: 40007865 PMCID: PMC11848209 DOI: 10.1039/d4cb00287c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Nucleic acids have the potential to form not only duplexes, but also various non-canonical secondary structures in living cells. Non-canonical structures play regulatory functions mainly in the central dogma. Therefore, nucleic acid targeting molecules are potential novel therapeutic drugs that can target 'undruggable' proteins in various diseases. One of the concerns of small molecules targeting nucleic acids is selectivity, because nucleic acids have only four different building blocks. Three- and four-stranded non-canonical structures, triplexes and quadruplexes, respectively, are promising targets of small molecules because their three-dimensional structures are significantly different from the canonical duplexes, which are the most abundant in cells. Here, we describe some basic properties of the triplexes and quadruplexes and small molecules targeting the triplexes and tetraplexes.
Collapse
Affiliation(s)
- Yoshiki Hashimoto
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Sumit Shil
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Mitsuki Tsuruta
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Keiko Kawauchi
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Daisuke Miyoshi
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| |
Collapse
|
17
|
Prabha S, Chauhan P, Warkare S, Pandey KM. A computational investigation of potential plant-based bioactive compounds against drug-resistant Staphylococcus aureus of multiple target proteins. J Biomol Struct Dyn 2025; 43:3311-3329. [PMID: 38133950 DOI: 10.1080/07391102.2023.2297009] [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: 06/09/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023]
Abstract
Drug-resistant Staphylococcus aureus (DRSA) poses a significant global health threat, like bacteremia, endocarditis, skin, soft tissue, bone, and joint infections. Nowadays, the resistance against conventional drugs has been a prompt and focused medical concern. The present study aimed to explore the inhibitory potential of plant-based bioactive compounds (PBBCs) against effective target proteins using a computational approach. We retrieved and verified 22 target proteins associated with DRSA and conducted a screening process that involved testing 87 PBBCs. Molecular docking was performed between screened PBBCs and reference drugs with selected target proteins via AutoDock. Subsequently, we filtered the target proteins and top PBBCs based on their binding affinity scores. Furthermore, molecular dynamic simulation was carried out through GROMACS for a duration of 100 ns, and the binding free energy was calculated using the gmx_MMPBSA. The result showed consistent hydrogen bonding interactions among the amino acid residues Ser 149, Arg 151, Thr 165, Thr 216, Glu 239, Ser 240, Ile 14, as well as Asn 18, Gln 19, Lys 45, Thr 46, Tyr 109, with their respective target proteins of the penicillin-binding protein and dihydrofolate reductase complex. Additionally, we assessed the pharmacokinetic properties of screened PBBCs via SwissADME and AdmetSAR. The findings suggest that β-amyrin, oleanolic acid, kaempferol, quercetin, and friedelin have the potential to inhibit the selected target proteins. In future research, both in vitro and in vivo, experiments will be needed to establish these PBBCs as potent antimicrobial drugs for DRSA.
Collapse
Affiliation(s)
- Sarit Prabha
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | | | - Sudeesh Warkare
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | - Khushhali M Pandey
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| |
Collapse
|
18
|
Lasorsa A, van der Wel PCA. Solid-state NMR protocols for unveiling dynamics and (drug) interactions of membrane-bound proteins. Protein Sci 2025; 34:e70102. [PMID: 40099898 PMCID: PMC11915643 DOI: 10.1002/pro.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Magic angle spinning solid-state NMR (MAS ssNMR) is a versatile tool for studying the structure and dynamics of membrane proteins, as well as their interactions with ligands and drugs. Its power lies in the ability to provide atomic-level information on samples under physiological-like conditions. Moreover, it can illuminate dynamics across a wide range of timescales with great relevance to membrane protein function and dysfunction. In this protocol paper, we highlight key aspects of sample preparation, data acquisition, and interpretation, based on our own experience and the broader literature. We discuss key protocol steps along with important considerations for sample preparation and parameters for ssNMR measurements, with reference to the special requirements of membrane-based samples. Such samples display physiologically relevant dynamics across different motional regimes that can be probed by NMR but also can interfere with certain NMR measurements. We guide the reader through the whole process from sample preparation to complex NMR characterization techniques. Throughout the report, we refer back to examples from our own prior work on the interactions between cytochrome c and cardiolipin-containing membranes, with a discussion of the lipid dependence and interactions with a peroxidase-activity inhibitor. We conclude with a short discussion of alternative and new methods that are further boosting the power and versatility of ssNMR as a tool to study membrane-bound proteins and their ligands or drug interactions.
Collapse
Affiliation(s)
- Alessia Lasorsa
- Zernike Institute for Advanced MaterialsUniversity of GroningenGroningenthe Netherlands
| | | |
Collapse
|
19
|
Inocencio I, Rai A, Donner D, Greening DW. The Proteomic Landscape of the Coronary Accessible Heart Cell Surfaceome. Proteomics 2025; 25:e202400320. [PMID: 39790063 PMCID: PMC11962585 DOI: 10.1002/pmic.202400320] [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: 10/02/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
Cell surface proteins (surfaceome) represent key signalling and interaction molecules for therapeutic targeting, biomarker profiling and cellular phenotyping in physiological and pathological states. Here, we employed coronary artery perfusion with membrane-impermeant biotin to label and capture the surface-accessible proteome in the neo-native (intact) heart. Using quantitative proteomics, we identified 701 heart cell surfaceome accessible by the coronary artery, including receptors, cell surface enzymes, adhesion and junctional molecules. This surfaceome comprises to 216 cardiac cell-specific surface proteins, including 29 proteins reported in cardiomyocytes (CXADR, CACNA1C), 12 in cardiac fibroblasts (ITGA8, COL3A1) and 63 in multiple cardiac cell types (ICAM1, SLC3A2, CDH2). Further, this surfaceome comprises to 53 proteins enriched in heart tissue compared to other tissues in humans and implicated in cardiac cell signalling networks involving cardiomyopathy (CDH2, DTNA, PTKP2, SNTA1, CAM, K2D/B), cardiac muscle contraction and development (ENG, SNTA1, SGCG, MYPN), calcium ion binding (SGCA, MASP1, THBS4, FBLN2, GSN) and cell metabolism (SDHA, NUDFS1, GYS1, ACO2, IDH2). This method offers a powerful tool for dissecting the molecular landscape of the coronary artery accessible heart cell surfaceome, its role in maintaining cardiac and vascular function, and potential molecular leads for studying cardiac cell interactions and systemic delivery to the neo-native heart.
Collapse
Affiliation(s)
- Iasmin Inocencio
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Baker Department of Cardiovascular Research Translation and ImplementationLa Trobe UniversityMelbourneVictoriaAustralia
| | - Alin Rai
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Baker Department of Cardiovascular Research Translation and ImplementationLa Trobe UniversityMelbourneVictoriaAustralia
- Baker Department of Cardiometabolic HealthUniversity of MelbourneMelbourneVictoriaAustralia
- Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Daniel Donner
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - David W. Greening
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Baker Department of Cardiovascular Research Translation and ImplementationLa Trobe UniversityMelbourneVictoriaAustralia
- Baker Department of Cardiometabolic HealthUniversity of MelbourneMelbourneVictoriaAustralia
- Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
20
|
Yang L, Liu M, Qi L, Liu Y, Lin X, Zhang YZ, Shen QT. Annealing synchronizes the TOM complex with Tom7 in a new orientation. Arch Biochem Biophys 2025; 766:110329. [PMID: 39924140 DOI: 10.1016/j.abb.2025.110329] [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: 10/26/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
Annealing is an ideal approach to synchronizing soluble proteins into their minimum-energy states via tandem heating and cooling treatments. Like soluble proteins, many membrane proteins also suffer intrinsic structural flexibility, the major obstacle to high-resolution structural determination. How to apply annealing onto membrane proteins remains unexplored. Here, we utilized the translocase of the outer mitochondrial membrane (TOM) as the model and investigated the ideal annealing conditions for membrane proteins. After structural determination via cryo-electron microscopy, we indicated that fast cooling the heated TOM complex to 0 °C can significantly improve the local resolution compared with the unannealed one. Structural analyses showed that annealing renders the TOM complex into a new conformation with its Tom7 α1 helix from a reclining position on the membrane surface to a lying orientation, accompanied by the loop between β6 and β7 in Tom40, flipping outward from the Tom40 β-barrel, ideal for preprotein translocation. In all, our results demonstrate the role of annealing in synchronizing membrane proteins and unveil unidentified conformations of the TOM complex.
Collapse
Affiliation(s)
- Liuyan Yang
- Marine Biotechnology Research Center, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China; School of Life Sciences, Department of Chemical Biology, Southern University of Science and Technology, Shenzhen, 518055, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Division of Healthy Oceans & Resource Sustainability, Laoshan Laboratory, Qingdao, 266237, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Mingdong Liu
- School of Life Sciences, Department of Chemical Biology, Southern University of Science and Technology, Shenzhen, 518055, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Division of Healthy Oceans & Resource Sustainability, Laoshan Laboratory, Qingdao, 266237, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lei Qi
- Biomedical Research Center for Structural Analysis, Shandong University, Jinan, 250012, China
| | - Yunhui Liu
- School of Life Sciences, Department of Chemical Biology, Southern University of Science and Technology, Shenzhen, 518055, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, 100191, China.
| | - Yu-Zhong Zhang
- Marine Biotechnology Research Center, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Qing-Tao Shen
- School of Life Sciences, Department of Chemical Biology, Southern University of Science and Technology, Shenzhen, 518055, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Division of Healthy Oceans & Resource Sustainability, Laoshan Laboratory, Qingdao, 266237, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
21
|
Amin H, Zamora-Ballesteros C, Diez-Casero JJ. Effects of Thermal and Antibiotic Treatments on the Viral Accumulation of FcMV1 in Fusarium circinatum Isolates. J Fungi (Basel) 2025; 11:267. [PMID: 40278088 PMCID: PMC12027980 DOI: 10.3390/jof11040267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Mycoviruses are viruses that infect fungi, including plant pathogens. The infection of these mycoviruses is sometimes associated with impaired phenotypes of their fungal hosts, a phenomenon known as hypovirulence. Thus, using mycoviruses as biological control agents has emerged as a promising tool to combat forest diseases. The invasive ascomycete fungus Fusarium circinatum, which causes pine pitch canker (PPC) disease in Pinus tree species and other coniferous trees, is infected by the mycovirus Fusarium circinatum mitovirus 1 (FcMV1), FcMV2-1, and FcMV2-2. However, its impact on pathogen fitness remains unclear. The most accurate method used to identify the effect of a mycovirus on its host is the generation of isogenic lines with and without the mycovirus. The present study aimed to cure F. circinatum isolates infected by FcMV1 using different approaches. For this purpose, three replicates of each isolate were exposed to thermal treatment (38 °C) and antibiotic treatment (ribavirin, cycloheximide, kanamycin, and rifampicin mixed with cAMP)(cyclic adenosine monophosphate) for five successive passages. The viral titer of FcMV1 was then assessed using qPCR (quantitative polymerase chain reaction) after the first week and after the fifth week of the treatment. The results revealed differences in treatment efficacy among F. circinatum isolates, with some showing very low virus titers at the end of the experiment. Both thermal and antibiotic treatment effectively reduced the viral load in all isolates. In addition, the antibiotic cycloheximide and rifampicin +cAMP reduced the viral titer more than ribavirin and kanamycin. The isolate Fc179 was found to be more prone to antibiotic treatment than the other two isolates (001 and Va221). This study demonstrated the possibility of using some isolates of F. circinatum for fine-tuning cures for mitovirus, in order to create virus-free strains for biological control in the future.
Collapse
Affiliation(s)
- Huma Amin
- Department of Plant Production and Forest Resources, Sustainable Forest Management Research Institute (iuFOR), Higher Technical School of Agricultural Engineering (ETSIIAA), University of Valladolid, 34004 Palencia, Spain
| | - Cristina Zamora-Ballesteros
- Faculty of Environment and Natural Resources, Albert-Ludwigs-Universität Freiburg, Bertoldstraße 17, 79098 Freiburg im Breisgau, Germany
| | - Julio Javier Diez-Casero
- Department of Plant Production and Forest Resources, Sustainable Forest Management Research Institute (iuFOR), Higher Technical School of Agricultural Engineering (ETSIIAA), University of Valladolid, 34004 Palencia, Spain
| |
Collapse
|
22
|
Vaz RF, Brown LS, Ladizhansky V. Membrane protein structure determination from Paramagnetic Relaxation Enhancement and internuclear distance restraints. JOURNAL OF BIOMOLECULAR NMR 2025:10.1007/s10858-025-00467-w. [PMID: 40156665 DOI: 10.1007/s10858-025-00467-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Magic angle spinning nuclear magnetic resonance (MAS NMR) is well suited for the determination of protein structure. The key structural information is obtained in the form of spectral cross peaks between spatially close nuclear spins, but assigning these cross peaks unambiguously to unique spin pairs is often a tedious task because of spectral overlap. Here, we use a seven-helical membrane protein Anabaena Sensory Rhodopsin (ASR) as a model system to demonstrate that transverse Paramagnetic Relaxation Enhancements (PRE) extracted from 2D MAS NMR spectra could be used to obtain a protein structural model. Starting with near complete assignments (93%) of ASR residues, TALOS + predicted backbone dihedral angles and secondary structure restraints in the form of backbone hydrogen bonds are combined with PRE-based restraints and used to generate a coarse model. This model is subsequently utilized as a template reference to facilitate automated assignments of highly ambiguous internuclear correlations. The template is used in an iterative cross peak assignment process and is progressively improved through the inclusion of disambiguated restraints, thereby converging to a low root-mean-square-deviation structural model. In addition to improving structure calculation conversion, the inclusion of PREs also improves packing between helices within an alpha-helical bundle.
Collapse
Affiliation(s)
- Raoul F Vaz
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Leonid S Brown
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Vlad Ladizhansky
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
23
|
Šolinc G, Srnko M, Merzel F, Crnković A, Kozorog M, Podobnik M, Anderluh G. Cryo-EM structures of a protein pore reveal a cluster of cholesterol molecules and diverse roles of membrane lipids. Nat Commun 2025; 16:2972. [PMID: 40140423 PMCID: PMC11947440 DOI: 10.1038/s41467-025-58334-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The structure and function of membrane proteins depend on their interactions with lipids that constitute membranes. Actinoporins are α-pore-forming proteins that bind preferentially to sphingomyelin-containing membranes, where they oligomerize and form transmembrane pores. Through a comprehensive cryo-electron microscopic analysis of a pore formed by an actinoporin Fav from the coral Orbicella faveolata, we show that the octameric pore interacts with 112 lipids in the upper leaflet of the membrane, reveal the roles of lipids, and demonstrate that the actinoporin surface is suited for binding multiple receptor sphingomyelin molecules. When cholesterol is present in the membrane, it forms a cluster of four molecules associated with each protomer. Atomistic simulations support the structural data and reveal additional effects of the pore on the lipid membrane. These data reveal a complex network of protein-lipid and lipid-lipid interactions and an underrated role of lipids in the structure and function of transmembrane protein complexes.
Collapse
Affiliation(s)
- Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marija Srnko
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Franci Merzel
- Theory Department, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ana Crnković
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Mirijam Kozorog
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| |
Collapse
|
24
|
Wang J, Guo C, Wei X, Pu X, Zhao Y, Xu C, Wang W. GPCR Sense Communication Among Interaction Nematodes with Other Organisms. Int J Mol Sci 2025; 26:2822. [PMID: 40141464 PMCID: PMC11943259 DOI: 10.3390/ijms26062822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Interactions between species give rise to chemical pathways of communication that regulate the interactions of transboundary species. The communication between nematodes and other species primarily occurs through the regulation of chemicals, with key species including plants, insects, bacteria, and nematode-trapping fungi that are closely associated with nematodes. G protein-coupled receptors (GPCRs) play a crucial role in interspecies communication. Certain flp genes, which function as GPCRs, exert varying degrees of influence on how nematodes interact with other species. These receptors facilitate the transmission of corresponding signals, thereby completing the interactions between species. This paper introduces the interactions between nematodes and other species and discusses the role of GPCRs in these organisms, contributing to a deeper understanding of the impact and significance of GPCRs in cross-border regulation between nematodes and other species. Furthermore, it is essential to leverage GPCRs in efforts to control pests.
Collapse
Affiliation(s)
- Jie Wang
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
- Key Laboratory of Northwest Cultivated Land Conservation and Marginal Land Improvement Enterprises, Ministry of Agriculture and Rural Affairs, Delingha 817000, China
| | - Changying Guo
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
| | - Xiaoli Wei
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
- Key Laboratory of Northwest Cultivated Land Conservation and Marginal Land Improvement Enterprises, Ministry of Agriculture and Rural Affairs, Delingha 817000, China
| | - Xiaojian Pu
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
- Key Laboratory of Northwest Cultivated Land Conservation and Marginal Land Improvement Enterprises, Ministry of Agriculture and Rural Affairs, Delingha 817000, China
| | - Yuanyuan Zhao
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
- Key Laboratory of Northwest Cultivated Land Conservation and Marginal Land Improvement Enterprises, Ministry of Agriculture and Rural Affairs, Delingha 817000, China
| | - Chengti Xu
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
- Key Laboratory of Northwest Cultivated Land Conservation and Marginal Land Improvement Enterprises, Ministry of Agriculture and Rural Affairs, Delingha 817000, China
| | - Wei Wang
- Academy of Animal Science and Veterinary, Qinghai University, Xining 810016, China; (J.W.); (C.G.); (X.W.); (X.P.); (Y.Z.)
- Key Laboratory of Northwest Cultivated Land Conservation and Marginal Land Improvement Enterprises, Ministry of Agriculture and Rural Affairs, Delingha 817000, China
| |
Collapse
|
25
|
Chatterjee H, Dutta P, Zacharias M, Sengupta N. Learning transition path and membrane topological signatures in the folding pathway of bacteriorhodopsin (BR) fragment with artificial intelligence. J Chem Phys 2025; 162:104110. [PMID: 40067008 DOI: 10.1063/5.0250082] [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: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 05/13/2025] Open
Abstract
Membrane protein folding in the viscous microenvironment of a lipid bilayer is an inherently slow process that challenges experiments and computational efforts alike. The folding kinetics is moreover associated with topological modulations of the biological milieu. Studying such structural changes in membrane-embedded proteins and understanding the associated topological signatures in membrane leaflets, therefore, remain relatively unexplored. Herein, we first aim to estimate the free energy barrier and the minimum free energy path (MFEP) connecting the membrane-embedded fully and partially inserted states of the bacteriorhodopsin fragment. To achieve this, we have considered independent sets of simulations from membrane-mimicking and membrane-embedded environments, respectively. An autoencoder model is used to elicit state-distinguishable collective variables for the system utilizing membrane-mimicking simulations. Our in-house Expectation Maximized Molecular Dynamics algorithm is initially used to deduce the barrier height between the two membrane-embedded states. Next, we develop the Geometry Optimized Local Direction search as a post-processing algorithm to identify the MFEP and the corresponding peptide conformations from the autoencoder-projected trajectories. Finally, we apply a graph attention neural network (GAT) model to learn the membrane surface topology as a function of the associated peptide structure, supervised by the membrane-embedded simulations. The resultant GAT model is then utilized to predict the membrane leaflet topology for the peptide structures along MFEP, obtained from membrane-mimicking simulations. The combined framework is expected to be useful in capturing key phenomena accompanying folding transitions in membranes. We discuss opportunities and avenues for further development.
Collapse
Affiliation(s)
- Hindol Chatterjee
- Department of Biological Science, Indian Institute of Science Education and Research (IISER), Kolkata, India
| | - Pallab Dutta
- Department of Biological Science, Indian Institute of Science Education and Research (IISER), Kolkata, India
| | - Martin Zacharias
- Technical University Munich, Ernst-Otto-Fischer-Straße 8, 85748 Garching, Germany
| | - Neelanjana Sengupta
- Department of Biological Science, Indian Institute of Science Education and Research (IISER), Kolkata, India
| |
Collapse
|
26
|
Riaz R, Khan K, Aghayeva S, Uddin R. Combatting antibiotic resistance in Gardnerella vaginalis: A comparative in silico investigation for drug target identification. PLoS One 2025; 20:e0314465. [PMID: 40073044 DOI: 10.1371/journal.pone.0314465] [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: 02/24/2024] [Accepted: 11/11/2024] [Indexed: 03/14/2025] Open
Abstract
Gardnerella vaginalis is the most frequently identified bacterium in approximately 95% of bacterial vaginosis (BV) cases. This species often exhibits resistance to multiple antibiotics, posing challenges for treatment. Therefore, there is an urgent need to develop and explore alternative therapeutic strategies for managing bacterial vaginosis. The objective of this study was to identify virulence factors and potential drug targets against Gardnerella vaginalis by utilizing in silico methods, including subtractive and comparative genomics. These methods enabled the systematic comparison of genetic sequences to pinpoint specific features unique to G. vaginalis and crucial for its pathogenicity, which could then inform the development of targeted therapeutic strategies. The analysis of the pathogen's proteomic data aimed to identify proteins that fulfilled specific criteria. These included being non-homologous to human proteins, essential for bacterial survival, amenable to drug targeting, involved in virulence, and contributing to antibiotic resistance. Following these analyses and an extensive literature review, the phospho-2-dehydro-3-deoxyheptonate aldolase enzyme emerged as a promising drug target. To deepen our understanding of the biological function of the identified protein, comprehensive protein structural modeling, validation studies, and network topology analyses were conducted. The subsequent structural analysis, encompassing modeling, validation, and network topology assessment, is aimed at further characterizing the protein. Using a library of around 9,000 FDA-approved compounds from the DrugBank database, a virtual screening was conducted to identify potential compounds that could effectively target the proposed drug target. This approach facilitated the evaluation of existing drugs for their ability to inhibit the target, potentially offering an efficient pathway for developing new treatments against the pathogen. Leveraging the established efficacy, safety, pharmacokinetics, and pharmacodynamics of these compounds, the study suggests repurposing them for Gardnerella vaginalis infections. Among the screened compounds, five specific agents-DB03332, DB07452, DB01262, DB02076, and DB00727-were identified as cost-effective therapeutic options for treating infections related to Gardnerella vaginalis. These compounds were selected based on their efficacy in targeting the pathogen while maintaining economic feasibility. While the results indicate potential efficacy in treating infections caused by the pathogen, further experimental studies are essential to validate these findings.
Collapse
Affiliation(s)
- Rabbia Riaz
- Baqai Institute of Information Technology, Baqai Medical University, Karachi, Pakistan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Kanwal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
27
|
Du Y, Cooper HJ. Native ambient mass spectrometry of membrane proteins directly from bacterial colonies. Chem Commun (Camb) 2025; 61:4168-4171. [PMID: 39963858 PMCID: PMC11833770 DOI: 10.1039/d4cc03881a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Native ambient mass spectrometry (NAMS) enables analysis of protein structure directly from biological substrates by use of liquid junction sampling techniques together with sampling solvents which mimic the proteins' natural environment. Here, we demonstrate detection of membrane and membrane-associated proteins directly from E. coli by combining liquid extraction surface analysis (LESA) with a straightforward washing protocol, which attenuates soluble proteins and enables detection of membrane proteins.
Collapse
Affiliation(s)
- Yuying Du
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Helen J Cooper
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
28
|
Knetsch TGJ, van Son H, Kobayashi M, Ubbink M. The Nature of Nanodisc Lipids Influences Fragment-Based Drug Discovery Results. Chem Biol Drug Des 2025; 105:e70080. [PMID: 40087816 PMCID: PMC11909325 DOI: 10.1111/cbdd.70080] [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: 10/02/2024] [Revised: 01/24/2025] [Accepted: 02/18/2025] [Indexed: 03/17/2025]
Abstract
Membrane proteins (MPs) are important yet challenging targets for drug discovery. MPs can be reconstituted in protein-lipid Nanodiscs (NDs), which resemble the native membrane environment. Drug-membrane interactions can affect the apparent binding stoichiometry and affinity, as well as the kinetics of ligands for a particular target, which is important for the extrapolation to pharmacokinetic studies. To investigate the role of the membrane, we have applied fragment-based drug discovery (FBDD) methods to cytochrome P450 3A4 (CYP3A4), reconstituted in NDs composed of different phosphocholine lipids: 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), dipalmitoylphosphatidylcholine (DPPC), or 1,2-diphytanoyl-sn-glycero-3-phosphocholine (DPhPC). Surface plasmon resonance screening of fragments and marketed drugs revealed extensive binding to the empty ND, correlating with analyte hydrophobicity, and the binding was critically dependent on ND lipid composition. POPC NDs showed much higher binding of fragments than DMPC and DPhPC NDs, resulting in a lower hit rate for CYP3A4 in POPC NDs, which demonstrated that the choice of the ND lipid is crucial to the outcome of a screen. The number of binders that were rejected based on atypical binding kinetics was lower for monomeric CYP3A4 in NDs than for non-native oligomeric CYP3A4 without the ND. Several fragments were exclusively identified as hits for CYP3A4 in the presence of the ND membrane. It is concluded that the nature of the ND is a critical factor for fragment screening of membrane proteins.
Collapse
Affiliation(s)
| | | | | | - Marcellus Ubbink
- Leiden Institute of ChemistryLeiden UniversityLeidenthe Netherlands
| |
Collapse
|
29
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
30
|
Hu W, Luo J, Li H, Luo Y, Zhang X, Wu Z, Yang Q, Zhao S, Hu B, Zou X. Identification of Key Genes in Esketamine's Therapeutic Effects on Perioperative Neurocognitive Disorders via Transcriptome Sequencing. Drug Des Devel Ther 2025; 19:981-1000. [PMID: 39974608 PMCID: PMC11836629 DOI: 10.2147/dddt.s510752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Background Esketamine ameliorates propofol-induced brain damage and cognitive impairment in mice. However, the precise role and underlying mechanism of esketamine in perioperative neurocognitive disorders (PND) remain unclear. Therefore, this study aimed to investigate the key genes associated with the role of esketamine in PND through animal modeling and transcriptome sequencing. Methods The present study established a mice model of PND and administered esketamine intervention to the model, and mice were divided into control, surgical group, and surgical group with esketamine. Behavioral assessments were conducted using the Morris water maze and Y maze paradigms, while transcriptome sequencing was performed on hippocampal samples obtained from 3 groups. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were performed on sequencing data to identify candidate genes related to esketamine treating PND. Thereafter, protein-protein interaction (PPI) network analysis was implemented to select key genes. The genes obtained from each step were subjected to enrichment analysis, and a regulatory network for key genes was constructed. Results The Morris water maze and Y maze findings demonstrated the successful construction of our PND model, and indicated that esketamine exhibits a certain therapeutic efficacy for PND. Ank1, Cbln4, L1cam, Gap43, and Shh were designated as key genes for subsequent analysis. The 5 key genes were significantly enriched in cholesterol biosynthesis, nonsense mediated decay (NMD), formation of a pool of free 40s subunits, major pathway of rRNA processing in the nucleolus and cytosol, among others. Notably, the miRNAs, mmu-mir-155-5p and mmu-mir-1a-3p, functionally co-regulated the expression of Ank1, Gap43, and L1cam. Conclusion We uncovered the therapeutic efficacy of esketamine in treating PND and identified 5 key genes (Ank1, Cbln4, L1cam, Gap43, and Shh) that contribute to its therapeutic effects, providing a valuable reference for further mechanistic studies on esketamine's treatment of PND.
Collapse
Affiliation(s)
- Wen Hu
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Jieqiong Luo
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Hui Li
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Yushan Luo
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Xiaoyuan Zhang
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Zhen Wu
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Qian Yang
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Sirun Zhao
- Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Bailong Hu
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| | - Xiaohua Zou
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People’s Republic of China
| |
Collapse
|
31
|
Sarcinella MC, Jones JD, Sorensen MJ, Edgcombe SA, Ruotolo BT, Kennedy RT, Bailey RC. Lipid Curvature and Fluidity Influence Lipid Incorporation Disparities in Nanodiscs. Anal Chem 2025; 97:2883-2889. [PMID: 39885642 DOI: 10.1021/acs.analchem.4c05494] [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: 02/01/2025]
Abstract
Nanodiscs have become a popular membrane mimetic system offering a well-defined bilayer environment to stabilize membrane proteins for in vitro analyses using a range of analytical methods; however, lipid compositions common to their deployment are simplistic and often fail to model native membrane complexity. Furthermore, there has been a general lack of rigorous analytical and biophysical characterization of nanodiscs comprising more than one lipid. To address these challenges, we coupled a nanodisc formation and purification workflow with targeted LC-MS/MS analysis to quantify lipids in nanodiscs made with different compositions. We screened lipids with a variety of headgroups and acyl chains and found that lipids did not always incorporate into nanodiscs at expected levels. Disparities in lipid incorporation were found to increase upon the addition of lipids known to induce curvature or rigidity to the membrane. Additionally, we found that adding just one additional type of lipid to nanodiscs changes the particle diameter and dispersity compared to nanodiscs containing a single lipid. We also formed and characterized nanodiscs using a complex starting composition inspired by the endoplasmic reticulum membrane and observed native-like cholesterol dynamics that modulated the lipid fluidity in the model bilayer system. Taken together, this work serves as a foundation for understanding nonstoichiometric lipid incorporation into nanodiscs and provides a basis for more thorough nanodisc characterization and quality control, which is critical to ensure multilipid nanodiscs synthesized accurately model the biological system of interest, enabling robust characterization of how the lipid landscape affects membrane protein structure and activity.
Collapse
Affiliation(s)
- Marina C Sarcinella
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Joshua D Jones
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Matthew J Sorensen
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Samantha A Edgcombe
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Brandon T Ruotolo
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Ryan C Bailey
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
32
|
Lutomski CA, Bennett JL, El-Baba TJ, Wu D, Hinkle JD, Burnap SA, Liko I, Mullen C, Syka JEP, Struwe WB, Robinson CV. Defining proteoform-specific interactions for drug targeting in a native cell signalling environment. Nat Chem 2025; 17:204-214. [PMID: 39806141 PMCID: PMC11794133 DOI: 10.1038/s41557-024-01711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Understanding the dynamics of membrane protein-ligand interactions within a native lipid bilayer is a major goal for drug discovery. Typically, cell-based assays are used, however, they are often blind to the effects of protein modifications. In this study, using the archetypal G protein-coupled receptor rhodopsin, we found that the receptor and its effectors can be released directly from retina rod disc membranes using infrared irradiation in a mass spectrometer. Subsequent isolation and dissociation by infrared multiphoton dissociation enabled the sequencing of individual retina proteoforms. Specifically, we categorized distinct proteoforms of rhodopsin, localized labile palmitoylations, discovered a Gβγ proteoform that abolishes membrane association and defined lipid modifications on G proteins that influence their assembly. Given reports of undesirable side-effects involving vision, we characterized the off-target drug binding of two phosphodiesterase 5 inhibitors, vardenafil and sildenafil, to the retina rod phosphodiesterase 6 (PDE6). The results demonstrate differential off-target reactivity with PDE6 and an interaction preference for lipidated proteoforms of G proteins. In summary, this study highlights the opportunities for probing proteoform-ligand interactions within natural membrane environments.
Collapse
Affiliation(s)
- Corinne A Lutomski
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jack L Bennett
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Tarick J El-Baba
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Di Wu
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | | | - Sean A Burnap
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | | | | | - Weston B Struwe
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Carol V Robinson
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Shah A, Wort JL, Ma Y, Pliotas C. Enabling structural biological electron paramagnetic resonance spectroscopy in membrane proteins through spin labelling. Curr Opin Chem Biol 2025; 84:102564. [PMID: 39709893 DOI: 10.1016/j.cbpa.2024.102564] [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: 09/20/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
Pulsed dipolar electron paramagnetic resonance spectroscopy (PDS), combined with site-directed spin-labelling, represents a powerful tool for the investigation of biomacromolecules, emerging as a keystone approach in structural biology. Increasingly, PDS is applied to study highly complex integral membrane protein systems, such as mechanosensitive ion channels, transporters, G-protein coupled receptors, ion pumps, and outer membrane proteins elucidating their dynamics and revealing conformational ensembles. Indeed, PDS offers a platform to study intermediate or lowly-populated states that are otherwise invisible to other modern methods, such as X-ray crystallography, cryo-EM, and hydrogen-deuterium exchange-mass spectrometry. Importantly, advances in spin labelling strategies welcome a new era of membrane protein investigation under near-native or in-cell conditions. Here, we review recent integral membrane protein PDS applications, and highlight well-suited, emerging spin labelling strategies that show promise for future studies.
Collapse
Affiliation(s)
- Anokhi Shah
- BioEmPiRe Centre for Structural Biological EPR Spectroscopy, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Manchester Institute of Biotechnology, University of Manchester, Manchester M1 7DN, UK
| | - Joshua L Wort
- BioEmPiRe Centre for Structural Biological EPR Spectroscopy, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Manchester Institute of Biotechnology, University of Manchester, Manchester M1 7DN, UK
| | - Yue Ma
- BioEmPiRe Centre for Structural Biological EPR Spectroscopy, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Manchester Institute of Biotechnology, University of Manchester, Manchester M1 7DN, UK
| | - Christos Pliotas
- BioEmPiRe Centre for Structural Biological EPR Spectroscopy, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Manchester Institute of Biotechnology, University of Manchester, Manchester M1 7DN, UK.
| |
Collapse
|
34
|
Wen P, Huang H, Zhang R, Zheng H, Liang T, Zhuang C, Wu Q, Wang J, Liu F, Zhang K, Wu W, He K, Liu F, Li H, Gu Z. Coacervate vesicles assembled by liquid-liquid phase separation improve delivery of biopharmaceuticals. Nat Chem 2025; 17:279-288. [PMID: 39806140 DOI: 10.1038/s41557-024-01705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
Vesicles play critical roles in cellular materials storage and signal transportation, even in the formation of organelles and cells. Natural vesicles are composed of a lipid layer that forms a membrane for the enclosure of substances inside. Here we report a coacervate vesicle formed by the liquid-liquid phase separation of cholesterol-modified DNA and histones. Unlike a phospholipid-based membrane-bounded vesicle, a coacervate vesicle lacks a membrane structure on the surface and is organized with a high-density liquid layer and a water-filled cavity. Through a straightforward coacervation process, we demonstrate that various biological agents, including virus particles, mRNA, cytokines and peptides, can be innocuously and directly enriched in the liquid phase. In contrast to the droplet-like coacervates that are prone to aggregation challenges, coacervate vesicles display superior kinetic stability, positioning them as a versatile delivery vehicle for biopharmaceuticals. We validate that incorporating oncolytic viruses into these coacervate vesicles endows them with potent oncolytic efficacy and elicits robust anti-tumour immune responses in mouse models.
Collapse
Affiliation(s)
- Ping Wen
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, China Medical University, Ministry of Education, Shenyang, China
- Phase I Clinical Trials Center, The First Hospital, China Medical University, Shenyang, China
| | - Ruizhe Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, China Medical University, Ministry of Education, Shenyang, China
- Phase I Clinical Trials Center, The First Hospital, China Medical University, Shenyang, China
| | - Hanqi Zheng
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tingxizi Liang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chuyue Zhuang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qing Wu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Junxia Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ke Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaixin He
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, China Medical University, Ministry of Education, Shenyang, China.
- Phase I Clinical Trials Center, The First Hospital, China Medical University, Shenyang, China.
| | - Hongjun Li
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
35
|
Su YY, Huang HC, Lin YT, Chuang YF, Sheu SY, Lin CC. HEDDI-Net: heterogeneous network embedding for drug-disease association prediction and drug repurposing, with application to Alzheimer's disease. J Transl Med 2025; 23:57. [PMID: 39891114 PMCID: PMC11786366 DOI: 10.1186/s12967-024-05938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/03/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The traditional process of developing new drugs is time-consuming and often unsuccessful, making drug repurposing an appealing alternative due to its speed and safety. Graph neural networks (GCNs) have emerged as a leading approach for predicting drug-disease associations by integrating drug and disease-related networks with advanced deep learning algorithms. However, GCNs generally infer association probabilities only for existing drugs and diseases, requiring network re-establishment and retraining for novel entities. Additionally, these methods often struggle with sparse networks and fail to elucidate the biological mechanisms underlying newly predicted drugs. METHODS To address the limitations of traditional methods, we developed HEDDI-Net, a heterogeneous embedding architecture designed to accurately detect drug-disease associations while preserving the interpretability of biological mechanisms. HEDDI-Net integrates graph and shallow learning techniques to extract representative diseases and proteins, respectively. These representative diseases and proteins are used to embed the input features, which are then utilized in a multilayer perceptron for predicting drug-disease associations. RESULTS In experiments, HEDDI-Net achieves areas under the receiver operating characteristic curve of over 0.98, outperforming state-of-the-art methods. Rigorous recovery analyses reveal a median recovery rate of 73% for the top 100 diseases, demonstrating its efficacy in identifying novel target diseases for existing drugs, known as drug repurposing. A case study on Alzheimer's disease highlighted the model's practical applicability and interpretability, identifying potential drug candidates like Baclofen, Fluoxetine, Pentoxifylline and Phenytoin. Notably, over 40% of the predicted candidates in the clusters of commonly prescribed clinical drugs Donepezil and Galantamine had been tested in clinical trials, validating the model's predictive accuracy and practical relevance. CONCLUSIONS HEDDI-NET represents a significant advancement by allowing direct application to new diseases and drugs without the need for retraining, a limitation of most GCN-based methods. Furthermore, HEDDI-Net provides detailed affinity patterns with representative proteins for predicted candidate drugs, facilitating an understanding of their physiological effects. This capability also supports the design and testing of alternative drugs that are similar to existing medications, enhancing the reliability and interpretability of potential repurposed drugs. The case study on Alzheimer's disease further underscores HEDDI-Net's ability to predict promising drugs and its applicability in drug repurposing.
Collapse
Affiliation(s)
- Yin-Yuan Su
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Ting Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Fang Chuang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Sheh-Yi Sheu
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Life Science and Institute of Genome Science, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
36
|
Medica S, Denton M, Diggins NL, Kramer-Hansen O, Crawford LB, Mayo AT, Perez WD, Daily MA, Parkins CJ, Slind LE, Pung LJ, Weber WC, Jaeger HK, Streblow ZJ, Sulgey G, Kreklywich CN, Alexander T, Rosenkilde MM, Caposio P, Hancock MH, Streblow DN. Third intracellular loop of HCMV US28 is necessary for signaling and viral reactivation. J Virol 2025; 99:e0180124. [PMID: 39655954 PMCID: PMC11784217 DOI: 10.1128/jvi.01801-24] [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: 10/22/2024] [Accepted: 11/13/2024] [Indexed: 02/01/2025] Open
Abstract
The human cytomegalovirus (HCMV) encoded chemokine receptor US28 plays a critical role in viral pathogenesis, mediating several processes such as cellular migration, differentiation, transformation, and viral latency and reactivation. Despite significant research examining the signal transduction pathways utilized by US28, the precise mechanism by which US28 activates these pathways remains unclear. We performed a mutational analysis of US28 to identify signaling domains that are critical for functional activities. Our results indicate that specific residues within the third intracellular loop (ICL3) of US28 are major determinants of G-protein coupling and downstream signaling activity. Alanine substitutions at positions S218, K223, and R225 attenuated US28-mediated activation of MAPK and RhoA signal transduction pathways. Furthermore, we show that mutations at positions S218, K223, or R225 result in impaired coupling to multiple Gα isoforms. However, these substitutions did not affect US28 plasma membrane localization or the receptor internalization rate. Utilizing CD34+ HPC models, we demonstrate that attenuation of US28 signaling via mutation of residues within the ICL3 region results in an inability of the virus to efficiently reactivate from latency. These results were recapitulated in vivo, utilizing a humanized mouse model of HCMV infection. Together, our results provide new insights into the mechanism by which US28 manipulates host signaling networks to mediate viral latency and reactivation. The results reported here will guide the development of targeted therapies to prevent HCMV-associated disease.IMPORTANCEHuman cytomegalovirus (HCMV) is a β-herpesvirus that infects between 44% and 100% of the world population. Primary infection is typically asymptomatic and results in the establishment of latent infection within CD34+hematopoietic progenitor cells (HPCs). However, reactivation from latent infection remains a significant cause of morbidity and mortality in immunocompromised individuals. The viral chemokine receptor US28 influences various cellular processes crucial for viral latency and reactivation, yet the precise mechanism by which US28 functions remains unclear. Through mutational analysis, we identified key residues within the third intracellular loop (ICL3) of US28 that govern G-protein coupling, downstream signaling, and viral reactivation in vitro and in vivo. These findings offer novel insights into how US28 manipulates host signaling networks to regulate HCMV latency and reactivation and expand our understanding of HCMV pathogenesis.
Collapse
Affiliation(s)
- Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Nicole L. Diggins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Olivia Kramer-Hansen
- Department of Biomedical Sciences Molecular Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Lindsey B. Crawford
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Adam T. Mayo
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Wilma D. Perez
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Michael A. Daily
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Luke E. Slind
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Lydia J. Pung
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Hannah K. Jaeger
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Zachary J. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Gauthami Sulgey
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Timothy Alexander
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Mette M. Rosenkilde
- Department of Biomedical Sciences Molecular Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Meaghan H. Hancock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, USA
| |
Collapse
|
37
|
Ofodile CA, Uzochukwu IC, Ezebuo FC, Ejiofor I, Adebola M, Okpoli I, Cubitt B, Witwit H, Okwuanaso CB, Onyemelukwe N, de la Torre JC. Flunarizine as a Candidate for Drug Repurposing Against Human Pathogenic Mammarenaviruses. Viruses 2025; 17:117. [PMID: 39861906 PMCID: PMC11768584 DOI: 10.3390/v17010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Lassa fever (LF), a viral hemorrhagic fever disease with a case fatality rate that can be over 20% among hospitalized LF patients, is endemic to many West African countries. Currently, no vaccines or therapies are specifically licensed to prevent or treat LF, hence the significance of developing therapeutics against the mammarenavirus Lassa virus (LASV), the causative agent of LF. We used in silico docking approaches to investigate the binding affinities of 2015 existing drugs to LASV proteins known to play critical roles in the formation and activity of the virus ribonucleoprotein complex (vRNP) responsible for directing replication and transcription of the viral genome. Validation of docking protocols were achieved with reference inhibitors of the respective targets. Our in silico docking screen identified five drugs (dexamethasone, tadalafil, mefloquine, ergocalciferol, and flunarizine) with strong predicted binding affinity to LASV proteins involved in the formation of the vRNP. We used cell-based functional assays to evaluate the antiviral activity of the five selected drugs. We found that flunarizine, a calcium-entry blocker, inhibited the vRNP activity of LASV and LCMV and virus surface glycoprotein fusion activity required for mammarenavirus cell entry. Consistently with these findings, flunarizine significantly reduced peak titers of LCMV in a multi-step growth kinetics assay in human A549 cells. Flunarizine is being used in several countries worldwide to treat vertigo and migraine, supporting the interest in exploring its repurposing as a candidate drug to treat LASV infections.
Collapse
Affiliation(s)
- Chukwudi A. Ofodile
- Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria; (C.A.O.); (C.B.O.)
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA; (B.C.); (H.W.)
- Department of Medical Laboratory Sciences, Faculty of Health Sciences and Technology, University of Nigeria, Nsukka 401105, Enugu, Nigeria;
| | - Ikemefuna C. Uzochukwu
- Department of Pharmaceutical & Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria; (I.C.U.); (F.C.E.); (I.O.)
| | - Fortunatus C. Ezebuo
- Department of Pharmaceutical & Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria; (I.C.U.); (F.C.E.); (I.O.)
- Department of Biochemistry, Graduate Center, City University of New York (CUNY), New York, NY 10016, USA
| | - InnocentMary Ejiofor
- Department of Pharmacognosy & Traditional Medicine, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria
| | - Mercy Adebola
- Department of Pharmaceutical & Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria; (I.C.U.); (F.C.E.); (I.O.)
| | - Innocent Okpoli
- Department of Pharmaceutical & Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria; (I.C.U.); (F.C.E.); (I.O.)
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA; (B.C.); (H.W.)
| | - Haydar Witwit
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA; (B.C.); (H.W.)
| | - Chetachi B. Okwuanaso
- Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, Nnamdi Azikiwe University, Awka 420218, Anambra, Nigeria; (C.A.O.); (C.B.O.)
| | - Ngozi Onyemelukwe
- Department of Medical Laboratory Sciences, Faculty of Health Sciences and Technology, University of Nigeria, Nsukka 401105, Enugu, Nigeria;
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA; (B.C.); (H.W.)
| |
Collapse
|
38
|
Fedorov II, Ivanov MV, Gorshkov MV. Effect of Drug-to-Protein Reaction Kinetics on the Results of Thermal Proteome Profiling. Anal Chem 2025; 97:22-26. [PMID: 39720990 DOI: 10.1021/acs.analchem.4c04313] [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: 12/26/2024]
Abstract
In this Letter, a two-term formalism for constructing protein solubility curves in thermal proteome profiling (TPP) is considered, which takes into account the efficiency of the drug-protein binding reaction. When the reaction is incomplete, this results in distortion of the otherwise sigmoidal shape of the curve after drug treatment, which is often observed in experiments. This distortion may be significant enough to disqualify the corresponding protein from the list of drug target candidates, thus negatively affecting the results of TPP data analysis. To further assist this analysis, we also developed the solubility curve simulation software to visualize the discussed effect. Several experimental data sets from recent TPP studies have been reprocessed, and we demonstrate in a few examples that the proposed two-term equation fits correctly the observed protein solubility curves with distorted shapes, also highlighting the previously unrecognized targets.
Collapse
Affiliation(s)
- Ivan I Fedorov
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Mark V Ivanov
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Mikhail V Gorshkov
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
39
|
Xu G, Yu J, Liu S, Cai L, Han XX. In situ surface-enhanced Raman spectroscopy for membrane protein analysis and sensing. Biosens Bioelectron 2025; 267:116819. [PMID: 39362137 DOI: 10.1016/j.bios.2024.116819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 09/08/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Membrane proteins are involved in a variety of dynamic cellular processes and exploration of the structural basis of membrane proteins is of significance for a better understanding of their functions. In situ analysis of membrane proteins and their dynamics is, however, challenging for conventional techniques. Surface-enhanced Raman spectroscopy (SERS) is powerful in protein structural characterization, allowing for sensitive, in-situ and real-time identification and dynamic monitoring under physiological conditions. In this review, the applications of SERS in probing membrane proteins are outlined, discussed and prospected. It starts with a brief introduction to membrane proteins, SERS theories and SERS-based strategies that commonly-used for membrane proteins. How to assemble phospholipid biolayers on SERS-active materials is highlighted, followed by respectively discussing about direct and indirect strategies for membrane protein sensing. SERS-based monitoring of protein-ligand interactions is finally introduced and its potential in biomedical applications is discussed in detail. The review ends with critical discussion about current challenges and limitations of this research field, and the promising perspectives in both fundamental and applied sciences.
Collapse
Affiliation(s)
- Guangyang Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Jiaheng Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Shiyi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, 130012, PR China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| |
Collapse
|
40
|
Oppelstrup T, Stanton LG, Tempkin JOB, Ozturk TN, Ingólfsson HI, Carpenter TS. Anisotropic interactions for continuum modeling of protein-membrane systems. J Chem Phys 2024; 161:244908. [PMID: 39786911 DOI: 10.1063/5.0237408] [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: 09/04/2024] [Accepted: 11/29/2024] [Indexed: 01/12/2025] Open
Abstract
In this work, a model for anisotropic interactions between proteins and cellular membranes is proposed for large-scale continuum simulations. The framework of the model is based on dynamic density functional theory, which provides a formalism to describe the lipid densities within the membrane as continuum fields while still maintaining the fidelity of the underlying molecular interactions. Within this framework, we extend recent results to include the anisotropic effects of protein-lipid interactions. As applications, we consider two membrane proteins of biological interest: a RAS-RAF complex tethered to the membrane and a membrane embedded G protein-coupled receptor. A strong qualitative and quantitative agreement is found between the numerical results and the corresponding molecular dynamics simulations. Combining the scope of continuum level simulations with the details from molecular level particle simulations enables research into protein-membrane behaviors at a more biologically relevant scale, which crucially can also be accessed via experiment.
Collapse
Affiliation(s)
- T Oppelstrup
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California 94550, USA
| | - L G Stanton
- Department of Mathematics and Statistics, San José State University, San José, California 95192, USA
| | - J O B Tempkin
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California 94550, USA
| | - T N Ozturk
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California 94550, USA
| | - H I Ingólfsson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California 94550, USA
| | - T S Carpenter
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California 94550, USA
| |
Collapse
|
41
|
Patel B, Patel S, Modi F, Patel A, Gelat B, Tanavde V, Vasavada A, Johar K. Combination of paclitaxel with rosiglitazone induces synergistic cytotoxic effects in ovarian cancer cells. Sci Rep 2024; 14:30672. [PMID: 39730440 DOI: 10.1038/s41598-024-74277-9] [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: 05/13/2024] [Accepted: 09/24/2024] [Indexed: 12/29/2024] Open
Abstract
Ovarian cancer is known to be a challenging disease to detect at an early stage and is a major cause of death among women. The current treatment for ovarian cancer typically involves a combination of surgery and the use of drugs such as platinum-based cytotoxic agents, anti-angiogenic drugs, etc. However, current treatment methods are not always effective in preventing the recurrence of ovarian cancer. As a result, the treatments administered after a relapse need to be more aggressive, leading to increased toxicity and drug resistance. To address this issue, researchers are exploring the potential of combining existing anticancer agents with novel or repurposed drugs to reduce the side effects and improve the effectiveness of treatment. In this study, we have investigated the use of rosiglitazone, a well-known anti-diabetic drug, in combination with the chemotherapeutic drug, paclitaxel for the prevention of ovarian cancer. The study utilized the SKOV-3 ovarian cancer cell line to assess the effects of this combination treatment. The results of the study showed that the combination of paclitaxel with rosiglitazone inhibited cell proliferation at much lower concentrations of paclitaxel as compared to paclitaxel alone. The combined treatment also induced cell cycle arrest at the G2/M phase and increased apoptosis by altering the mitochondrial membrane potential of the cells. Additionally, the combination treatment activated the PPAR-γ pathway and downregulated expression of genes associated with cancer stemness, such as NANOG, OCT4, and EHF. Furthermore, the CAM assay substantiated the anti-angiogenic potential of the synergistic treatment of paclitaxel and rosiglitazone. The findings of the study suggest that repurposing rosiglitazone as an anticancer agent in combination with paclitaxel has immense potential to target cancer cell cycle progression and apoptosis, making it a promising therapeutic approach for sensitizing chemo-resistant population of ovarian cancer cells.
Collapse
Affiliation(s)
- Binita Patel
- Department of Life Sciences, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Shanaya Patel
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Ahmedabad, 380009, Gujarat, India
| | - Foram Modi
- Department of Zoology, Biomedical Technology, Human Genetics, and WBC, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Aditi Patel
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Ahmedabad, 380009, Gujarat, India
| | - Brijesh Gelat
- Department of Zoology, Biomedical Technology, Human Genetics, and WBC, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Vivek Tanavde
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Ahmedabad, 380009, Gujarat, India
| | - Abhay Vasavada
- Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, 380052, Gujarat, India
| | - Kaid Johar
- Department of Zoology, Biomedical Technology, Human Genetics, and WBC, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India.
| |
Collapse
|
42
|
Denger A, Helms V. Identifying optimal substrate classes of membrane transporters. PLoS One 2024; 19:e0315330. [PMID: 39700222 DOI: 10.1371/journal.pone.0315330] [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: 06/24/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
Membrane transporters are responsible for moving a wide variety of molecules across biological membranes, making them integral to key biological pathways in all organisms. Identifying all membrane transporters within a (meta-)proteome, along with their specific substrates, provides important information for various research fields, including biotechnology, pharmacology, and metabolomics. Protein datasets are frequently annotated with thousands of molecular functions that form complex networks, often with partial or full redundancy and hierarchical relationships. This complexity, along with the low sample count for more specific functions, makes them unsuitable as classes for supervised learning methods, meaning that the creation of an optimal subset of annotations is required. However, selection of this subset requires extensive manual effort, along with knowledge about the biology behind the respective functions. Here, we present an automated pipeline to address this problem. Unlike previous approaches for reducing redundancy in GO datasets, we employ machine learning to identify a subset of functional annotations in a training dataset. Classes in the resulting predictive model meet four essential criteria: sufficient sample size for training predictive models, minimal redundancy, strong class separability, and relevance to substrate transport. Furthermore, we implemented a pipeline for creating training datasets of transmembrane transporters that cover a wide range of organisms, including plants, bacteria, mammals, and single-cell eukaryotes. For a dataset containing 98.1% of transporters from S. cerevisiae, the pipeline automatically reduced the number of functional annotations from 287 to 11 GO terms that could be classified with a median pairwise F1 score of 0.87±0.16. For a meta-organism dataset containing 96% of all transport proteins from S. cerevisiae, A. thaliana, E. coli and human, the number of classes was reduced from 695 to 49, with a median F1 score of 0.92±0.10 between pairs of GO terms. When lowering the percentage of covered proteins down to 67%, the pipeline found a subset of 30 GO terms with a median F1 score of 0.95±0.06.
Collapse
Affiliation(s)
- Andreas Denger
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
43
|
Voldřich J, Matoušová M, Šmídková M, Mertlíková‐Kaiserová H. Fluorescence-Based HTS Assays for Ion Channel Modulation in Drug Discovery Pipelines. ChemMedChem 2024; 19:e202400383. [PMID: 39221492 PMCID: PMC11648840 DOI: 10.1002/cmdc.202400383] [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: 05/20/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Ion channels represent a druggable family of transmembrane pore-forming proteins with important (patho)physiological functions. While electrophysiological measurement (manual patch clamp) remains the only direct method for detection of ion currents, it is a labor-intensive technique. Although automated patch clamp instruments have become available to date, their high costs limit their use to large pharma companies or commercial screening facilities. Therefore, fluorescence-based assays are particularly important for initial screening of compound libraries. Despite their numerous disadvantages, they are highly amenable to high-throughput screening and in many cases, no sophisticated instrumentation or materials are required. These features predispose them for implementation in early phases of drug discovery pipelines (hit identification), even in an academic environment. This review summarizes the advantages and pitfalls of individual methodological approaches for identification of ion channel modulators employing fluorescent probes (i. e., membrane potential and ion flux assays) with emphasis on practical aspects of their adaptation to high-throughput format.
Collapse
Affiliation(s)
- Jan Voldřich
- Institute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech RepublicFlemingovo nam. 2Prague 6 – Dejvice16610Czech Republic
- University of Chemistry and TechnologyTechnická 5Prague 6 – Dejvice166 28Czech Republic
| | - Marika Matoušová
- Institute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech RepublicFlemingovo nam. 2Prague 6 – Dejvice16610Czech Republic
| | - Markéta Šmídková
- Institute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech RepublicFlemingovo nam. 2Prague 6 – Dejvice16610Czech Republic
| | - Helena Mertlíková‐Kaiserová
- Institute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech RepublicFlemingovo nam. 2Prague 6 – Dejvice16610Czech Republic
| |
Collapse
|
44
|
Zia T, Khan K, Aghayeva S, Uddin R. Breaking resistance: in silico subtractive and comparative genomics approaches for drug targeting in Bacteroides fragilis. Biotechnol Lett 2024; 46:1249-1268. [PMID: 39424748 DOI: 10.1007/s10529-024-03537-5] [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: 06/10/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
The purpose of this study was to identify potential novel drug targets for Bacteroides fragilis infections using bioinformatics techniques, such as subtractive and comparative genomics. Bacteroides fragilis is a frequently isolated anaerobic pathogen, particularly in the human digestive tract, where its pathogenesis and persistence are influenced by various virulence factors. By understanding these factors, the study aims to explore alternative therapeutic strategies and provide insights for the development of treatments against B. fragilis infections, particularly as alternatives to antibiotic therapy. A comparative subtractive genomic analysis was performed against the B. fragilis (strain CL07T12C05) to identify unique drug targets. The analysis includes the identification of non-paralogous, non-homologous, essential, and drug target like proteins. Moreover, a comprehensive structural analysis of the protein was conducted utilizing structure modeling and validation techniques, along with network topology analysis. Furthermore, a library comprising approximately 9000 FDA-approved compounds accessible in the DrugBank database was employed to conduct virtual screenings for compounds effective against the designated drug target. The top shortlisted compounds were further studied by employing MD simulations using GROMACS. This approach was chosen due to the established safety, efficacy, pharmacokinetics, and toxicity profiles of these compounds. As a result, B. fragilis (strain CL07T12C05) was found to possess 4595 proteins. Among these, 3518 were identified as non-homologous, 1508 deemed essential for bacterial viability, 348 exhibited drug-like properties, 203 were implicated in virulence, and 135 displayed antibiotic resistance. Following an extensive literature review, the protein Sialic acid O-acetyltransferase was chosen through a hierarchical shortlisting process as a potential therapeutic target. The ongoing research facilitated the repurposing of drug compounds: DB12411, DB02112, DB03591, and DB00192, as cost-effective medications against B. fragilis related infections. MD simulations analysis showed that DB12411 may be a potential drug candidate against Sialic acid O-acetyltransferase from B. fragilis. Through subtractive and comparative genomic analysis, Sialic acid O-acetyltransferase was identified as a promising drug target against Bacteroides fragilis. The findings indicate that compounds targeting this protein could potentially be effective in treating B. fragilis infections. However, further experimental validation is required to conclusively confirm their efficacy.
Collapse
Affiliation(s)
- Tehreem Zia
- Baqai Institute of Information Technology, Baqai Medical University, Karachi, Pakistan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Kanwal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan.
| |
Collapse
|
45
|
Kleinau G, Chini B, Andersson L, Scheerer P. The role of G protein-coupled receptors and their ligands in animal domestication. Anim Genet 2024; 55:893-906. [PMID: 39324206 DOI: 10.1111/age.13476] [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: 09/11/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
The domestication of plants and animals has resulted in one of the most significant cultural and socio-economical transitions in human history. Domestication of animals, including human-supervised reproduction, largely uncoupled particular animal species from their natural, evolutionary history driven by environmental and ecological factors. The primary motivations for domesticating animals were, and still are, producing food and materials (e.g. meat, eggs, honey or milk products, wool, leather products, jewelry and medication products) to support plowing in agriculture or in transportation (e.g. horse, cattle, camel and llama) and to facilitate human activities (for hunting, rescuing, therapeutic aid, guarding behavior and protecting or just as a companion). In recent years, decoded genetic information from more than 40 domesticated animal species have become available; these studies have identified genes and mutations associated with specific physiological and behavioral traits contributing to the complex genetic background of animal domestication. These breeding-altered genomes provide insights into the regulation of different physiological areas, including information on links between e.g. endocrinology and behavior, with important pathophysiological implications (e.g. for obesity and cancer), extending the interest in domestication well beyond the field. Several genes that have undergone selection during domestication and breeding encode specific G protein-coupled receptors, a class of membrane-spanning receptors involved in the regulation of a number of overarching functions such as reproduction, development, body homeostasis, metabolism, stress responses, cognition, learning and memory. Here we summarize the available literature on variations in G protein-coupled receptors and their ligands and how these have contributed to animal domestication.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Bice Chini
- CNR, Institute of Neuroscience, Vedano al Lambro, Italy, and NeuroMI - Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Patrick Scheerer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| |
Collapse
|
46
|
Wang Y, Wang F, Liu W, Geng Y, Shi Y, Tian Y, Zhang B, Luo Y, Sun X. New drug discovery and development from natural products: Advances and strategies. Pharmacol Ther 2024; 264:108752. [PMID: 39557343 DOI: 10.1016/j.pharmthera.2024.108752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Natural products (NPs) have a long history as sources for drug discovery, more than half of approved drugs are related to NPs, which also exhibit multifaceted advantages in the clinical treatment of complex diseases. However, bioactivity screening of NPs, target identification, and design optimization require continuously improved strategies, the complexity of drug mechanism of action and the limitations of technological strategies pose numerous challenges to the development of new drugs. This review begins with an overview of bioactivity- and target-based drug development patterns for NPs, advances in NP screening and derivatization, and the advantages and problems of major targets such as genes and proteins. Then, target-based drugs as well as identification and validation methods are further discussed to elucidate their mechanism of action. Subsequently, the current status and development trend of the application of traditional and emerging technologies in drug discovery and development of NPs are systematically described. Finally, the collaborative strategy of multi-technology integration and multi-disciplinary intersection is emphasized for the challenges faced in the identification, optimization, activity evaluation, and clinical application of NPs. It is hoped to provide a systematic overview and inspiration for exploring new drugs from natural resources in the future.
Collapse
Affiliation(s)
- Yixin Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Fan Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Wenxiu Liu
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Yifei Geng
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Yahong Shi
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Yu Tian
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| |
Collapse
|
47
|
Huang C, Zhang J, Liu Z, Xu J, Zhao Y, Zhang P. In Situ and Label-Free Quantification of Membrane Protein-Ligand Interactions Using Optical Imaging Techniques: A Review. BIOSENSORS 2024; 14:537. [PMID: 39589996 PMCID: PMC11592237 DOI: 10.3390/bios14110537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024]
Abstract
Membrane proteins are crucial for various cellular processes and are key targets in pharmacological research. Their interactions with ligands are essential for elucidating cellular mechanisms and advancing drug development. To study these interactions without altering their functional properties in native environments, several advanced optical imaging methods have been developed for in situ and label-free quantification. This review focuses on recent optical imaging techniques such as surface plasmon resonance imaging (SPRi), surface plasmon resonance microscopy (SPRM), edge tracking approaches, and surface light scattering microscopy (SLSM). We explore the operational principles, recent advancements, and the scope of application of these methods. Additionally, we address the current challenges and explore the future potential of these innovative optical imaging strategies in deepening our understanding of biomolecular interactions and facilitating the discovery of new therapeutic agents.
Collapse
Affiliation(s)
- Caixin Huang
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jingbo Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaoyang Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiying Xu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Zhao
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Pengfei Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
48
|
Reddy AT, Lakshmi SP, Nyunoya T, Reddy RC. Comprehensive characterization of a novel small-molecule activator for the nuclear receptor Nur77: Chemical, molecular, and biological insights. Eur J Pharmacol 2024; 982:176945. [PMID: 39182547 DOI: 10.1016/j.ejphar.2024.176945] [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: 06/01/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
The nuclear receptor Nur77 is a transcription factor belonging to the NR4A subfamily. Upon activation, it regulates a wide array of biological and pathophysiological processes by modulating the expression of its target genes. Previous findings have classified Nur77 as an orphan receptor because of the discovery of a structurally atypical ligand-binding domain and the lack of identification of an endogenous ligand. Nevertheless, recent studies have uncovered several endogenous, natural, and small synthetic molecules that can bind to and activate Nur77. However, developing selective and potent Nur77 activators remains a significant challenge. The discovery of novel and potential small synthetic molecules that modulate Nur77 activity will facilitate therapeutic interventions of Nur77 against several human diseases. In this study, we have reported the development of a novel and effective Nur77 ligand. Our data show that (1E,4E)-1,5-di(pyrazin-2-yl)penta-1,4-dien-3-one (PB) induces Nur77 transcriptional activity by interacting with a putative Nur77 ligand binding site by forming solid hydrogen bonding. Calculated chemical parameters denote that PB has sophisticated chemical features that will enhance its interaction with the Nur77 ligand-binding domain. Molecular docking simulations showed that PB fits in the Nur77 putative ligand binding site with solid hydrogen bonding, and molecular dynamics simulations indicate that PB binding would stabilize the Nur77 ligand binding domain. Further, in vitro studies revealed that PB increases Nur77 nuclear expression and activity, inhibits cigarette smoke-induced inflammatory phenotype of airway epithelial cells, and protects against apoptosis. These findings provide insights into developing an effective Nur77 small-molecule activator which could be developed into a therapeutic agent against inflammatory diseases.
Collapse
Affiliation(s)
- Aravind T Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| | - Sowmya P Lakshmi
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Toru Nyunoya
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Raju C Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
49
|
Kwoji ID, Okpeku M, Aiyegoro OA, Adeleke MA. Metabolic interactions of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 in co-culture: implications for multi-strain probiotics. J Appl Microbiol 2024; 135:lxae264. [PMID: 39510973 DOI: 10.1093/jambio/lxae264] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
AIMS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 are potential probiotic bacteria. The mechanisms of enhanced benefits by muti-strain probiotics are yet fully understood. We elucidated the influence of co-culturing on the metabolite profiles of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 to decipher the impacts of co-culturing on metabolic interactions between the strains. METHODS AND RESULTS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 were grown in single and co-cultures in defined media. Bacterial cell metabolites were extracted at the mid-stationary growth phase and analysed using two-dimensional gas chromatography-time-of-flight mass spectrometry (GC × GC-TOFMS). Mass-spectral data were preprocessed and analysed using unsupervised and supervised methods based on the group allocations. A total of 1387 metabolites were identified, with 18.31% significant metabolites (P < 0.05) and 10.17% differential metabolites (P < 0.05, variable importance on projection > 1). The differential metabolites identified include arabinofuranose, methyl-galactoside, N-acetylglutamic acid, phosphoric acid, and decanoic acid. The metabolites impacted carbohydrate and amino-sugar metabolism. CONCLUSION Co-culturing of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 influenced the metabolite profiles of the strains and impacted metabolic/biosynthetic pathways, indicating cell-to-cell interactions between the strains.
Collapse
Affiliation(s)
- Iliya Dauda Kwoji
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Olayinka Ayobami Aiyegoro
- Unit for Environmental Sciences and Management, Northwest University, Potchefstroom, Northwest 2520, South Africa
| | - Matthew Adekunle Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| |
Collapse
|
50
|
Shen Z, Wang J, Chen Y, Fang P, Yuan A, Chen AF, Yan X, Lyu Y, Pu J. Activation of nuclear receptor pregnane-X-receptor protects against abdominal aortic aneurysm by inhibiting oxidative stress. Redox Biol 2024; 77:103397. [PMID: 39427444 PMCID: PMC11534186 DOI: 10.1016/j.redox.2024.103397] [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: 08/19/2024] [Revised: 10/05/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening condition, but effective medications to prevent its progression and rupture are currently lacking. The nuclear receptor pregnane-X-receptor (PXR) plays a crucial role in vascular homeostasis. However, the role of PXR in AAA development remains unknown. We first detected the PXR expression in human and murine AAA tissues by RT-qPCR and Western blot. To investigate the potential role of PXR in the development of AAA, we used adeno-associated virus-mediated overexpression of PXR and pharmacological activation of PXR by ginkgolide A (GA) in mouse AAA models induced by both angiotensin II (AngII) and calcium phosphate [Ca3(PO4)2]. The underlying mechanism was further explored using RNA-sequencing and molecular biological analyses. We found a significant decrease in both mRNA and protein levels of PXR in both human and murine aortic smooth muscle cells from AAA tissues, accompanied with phenotypic switching of vascular smooth muscle cell and increased oxidative stress. PXR overexpression in abdominal aortas and GA treatment successfully suppressed AAA formation in both mouse AAA models. RNA-sequencing data revealed that PXR activation inhibited gamma-aminobutyric acid type A receptor subunit alpha3 (GABRA3) expression. Additional mechanistic studies identified that PXR suppressed AAA through mitigating GABRA3-induced reactive oxygen species (ROS) generation and subsequent phosphorylation of c-Jun N-terminal kinase (JNK). Interestingly, p-JNK was found to induce ubiquitin-proteasome degradation of PXR. In summary, our data unveiled, for the first time, the protective role of PXR against AAA pathogenesis by inhibiting oxidative stress. These findings suggested PXR as a promising therapeutic target for AAA.
Collapse
MESH Headings
- Pregnane X Receptor/metabolism
- Pregnane X Receptor/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Oxidative Stress/drug effects
- Animals
- Humans
- Mice
- Disease Models, Animal
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
Collapse
Affiliation(s)
- Zhi Shen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Jinxi Wang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yifei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Peiliang Fang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Ancai Yuan
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuyan Lyu
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|