Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (541)

Search Parameters:
Keywords = toxin-binding protein

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 5546 KiB  
Proceeding Paper
RhsP2 Protein as a New Antibacterial Toxin Targeting RNA
by Tamara Nami Haj Marza
Med. Sci. Forum 2025, 35(1), 3; https://doi.org/10.3390/msf2025035003 - 24 Jul 2025
Abstract
Many bacteria, such as Pseudomonas aeruginosa, have encoded many toxins like RhsP2 that target non-coding RNAs (ncRNAs) in a similar mechanism to ART components; bacterial RNA loses its function of amino acid translation. A virtual screening approach was used to investigate RhsP2, [...] Read more.
Many bacteria, such as Pseudomonas aeruginosa, have encoded many toxins like RhsP2 that target non-coding RNAs (ncRNAs) in a similar mechanism to ART components; bacterial RNA loses its function of amino acid translation. A virtual screening approach was used to investigate RhsP2, which targets 16s rRNAs and then disrupts the translation of bacterial amino acids to proteins. Rifamycin is the bioreference as it forms a stable complex with the bacterial RNA in its active sites. Using different docking software can determine the best predicted conformations between RhsP2/16S and rRNA, and analyzing the docking score for both Affinity Binding and the root mean square deviation (RMSD) of particle coordinates helps choose the most appropriate drugs by using tools such as bioinformatics platforms and databases. Full article
(This article belongs to the Proceedings of The 4th International Electronic Conference on Antibiotics)
Show Figures

Figure 1

21 pages, 6401 KiB  
Article
The Dissociation of Latrophilin Fragments by Perfluorooctanoic Acid (PFOA) Inhibits LTXN4C-Induced Neurotransmitter Release
by Evelina Petitto, Jennifer K. Blackburn, M. Atiqur Rahman and Yuri A. Ushkaryov
Toxins 2025, 17(7), 359; https://doi.org/10.3390/toxins17070359 - 20 Jul 2025
Viewed by 209
Abstract
α-Latrotoxin stimulates neurotransmitter release by binding to a presynaptic receptor and then forming ion-permeable membrane pores and/or stimulating the receptor, latrophilin-1, or Adhesion G-protein-coupled receptor type L1 (ADGRL1). To avoid pore formation, we use the mutant α-latrotoxin (LTXN4C), which does not [...] Read more.
α-Latrotoxin stimulates neurotransmitter release by binding to a presynaptic receptor and then forming ion-permeable membrane pores and/or stimulating the receptor, latrophilin-1, or Adhesion G-protein-coupled receptor type L1 (ADGRL1). To avoid pore formation, we use the mutant α-latrotoxin (LTXN4C), which does not form pores and only acts through ADGRL1. ADGRL1 is cleaved into an N-terminal fragment (NTF) and a C-terminal fragment (CTF), which behave as independent cell-surface proteins, reassociating upon binding LTXN4C. We investigated the role of the NTF-CTF association in LTXN4C action, using perfluorooctanoic acid (PFOA). We demonstrate that at low concentrations (≤100 μM) PFOA does not adversely affect ADGRL1-expressing neuroblastoma cells or inhibit LTXN4C binding. However, it causes the dissociation of the NTF-CTF complexes, independent redistribution of the fragments on the cell surface, and their separate internalization. PFOA also promotes the dissociation of NTF-CTF complexes induced by LTXN4C binding. When applied to mouse neuromuscular junctions, PFOA inhibits LTXN4C-induced neurotransmitter release in a concentration-dependent manner. Our results indicate that ADGRL1 can mediate LTXN4C signaling only while its fragments remain associated. These findings explain some aspects of receptor-dependent toxin action and contribute to a mechanistic understanding of ADGRL1 functions in neurons. Full article
Show Figures

Graphical abstract

13 pages, 3226 KiB  
Article
Nematocidal Activity and Intestinal Receptor-Binding Affinity of Endogenous Lectins in Bursaphelenchus xylophilus (Pinewood Nematode)
by Songqing Wu, Yunzhu Sun, Zibo Li, Xinquan Li, Wei Yu and Yajie Guo
Forests 2025, 16(7), 1177; https://doi.org/10.3390/f16071177 - 16 Jul 2025
Viewed by 229
Abstract
Pine wilt disease, a devastating disease severely impacting pine ecosystems, is caused by the pinewood nematode Bursaphelenchus xylophilus (Steiner & Bührer, 1934) Nickle, 1970 (Nematoda: Parasitaphelenchidae). Controlling B. xylophilus is crucial for preventing and managing pine wilt disease. Recently discovered novel nematocidal lectins [...] Read more.
Pine wilt disease, a devastating disease severely impacting pine ecosystems, is caused by the pinewood nematode Bursaphelenchus xylophilus (Steiner & Bührer, 1934) Nickle, 1970 (Nematoda: Parasitaphelenchidae). Controlling B. xylophilus is crucial for preventing and managing pine wilt disease. Recently discovered novel nematocidal lectins could provide more advantageous materials for utilizing genetically engineered bacteria to control this pathogen. Therefore, this study focuses on identifying novel nematocidal toxins within B. xylophilus lectins. Overall, we obtained twenty-one galectin, one L-type lectin (LTL), and three chitin-binding domain (CBD) genes by screening the B. xylophilus genome database; these genes were successfully expressed proteins. The bioassay results indicated that Bxgalectin2, Bxgalectin3, Bxgalectin4, Bxgalectin9, and BxLTL1 induced mortality rates exceeding 50% in B. xylophilus. Notably, Bxgalectin4 showed the strongest nematocidal activity, causing 88% mortality in the treated nematode population. The enzyme-linked immunosorbent assays further demonstrated that Bxgalectin3 (Kd = 8.992 nM) and Bxgalectin4 (Kd = 9.634 nM) had a higher binding affinity to GPI-anchored proteins from B. xylophilus. Additionally, Bxgalectin2 (Kd = 16.50 nM), Bxgalectin9 (Kd = 16.48 nM), and BxLTL1 (Kd = 24.34 nM) can bind to the GPI-anchored protein. This study reports, for the first time, that lectins endogenous to B. xylophilus exhibit nematocidal activity against their own species. These findings open up the possibility of using nematode lectins as potent control agents in the biological control of B. xylophilus. Full article
(This article belongs to the Section Forest Health)
Show Figures

Figure 1

27 pages, 3232 KiB  
Article
Genomic and Functional Characterization of Multidrug-Resistant E. coli: Insights into Resistome, Virulome, and Signaling Systems
by Vijaya Bharathi Srinivasan, Naveenraj Rajasekar, Karthikeyan Krishnan, Mahesh Kumar, Chankit Giri, Balvinder Singh and Govindan Rajamohan
Antibiotics 2025, 14(7), 667; https://doi.org/10.3390/antibiotics14070667 - 30 Jun 2025
Viewed by 399
Abstract
Introduction: Genetic plasticity and adaptive camouflage in critical pathogens have contributed to the global surge in multidrug-resistant (MDR) infections, posing a serious threat to public health and therapeutic efficacy. Antimicrobial resistance, now a leading cause of global mortality, demands urgent action through diagnostics, [...] Read more.
Introduction: Genetic plasticity and adaptive camouflage in critical pathogens have contributed to the global surge in multidrug-resistant (MDR) infections, posing a serious threat to public health and therapeutic efficacy. Antimicrobial resistance, now a leading cause of global mortality, demands urgent action through diagnostics, vaccines, and therapeutics. In India, the Indian Council of Medical Research’s surveillance network identifies Escherichia coli as a major cause of urinary tract infections, with increasing prevalence in human gut microbiomes, highlighting its significance across One Health domains. Methods: Whole-genome sequencing of E. coli strain ECG015, isolated from a human gut sample, was performed using the Illumina NextSeq platform. Results: Genomic analysis revealed multiple antibiotic resistance genes, virulence factors, and efflux pump components. Phylogenomic comparisons showed close relatedness to pathovars from both human and animal origins. Notably the genome encoded protein tyrosine kinases (Etk/Ptk and Wzc) and displayed variations in the envelope stress-responsive CpxAR two-component system. Promoter analysis identified putative CpxR-binding sites upstream of genes involved in resistance, efflux, protein kinases, and the MazEF toxin–antitoxin module, suggesting a potential regulatory role of CpxAR in stress response and persistence. Conclusions: This study presents a comprehensive genomic profile of E. coli ECG015, a gut-derived isolate exhibiting clinically significant resistance traits. For the first time, it implicates the CpxAR two-component system as a potential central regulator coordinating antimicrobial resistance, stress kinase signaling, and programmed cell death. These findings lay the groundwork for future functional studies aimed at targeting stress-response pathways as novel intervention strategies against antimicrobial resistance. Full article
(This article belongs to the Special Issue Genomic Analysis of Drug-Resistant Pathogens)
Show Figures

Figure 1

23 pages, 1423 KiB  
Review
Transporter-Mediated Interactions Between Uremic Toxins and Drugs: A Hidden Driver of Toxicity in Chronic Kidney Disease
by Pierre Spicher, François Brazier, Solène M. Laville, Sophie Liabeuf, Saïd Kamel, Maxime Culot and Sandra Bodeau
Int. J. Mol. Sci. 2025, 26(13), 6328; https://doi.org/10.3390/ijms26136328 - 30 Jun 2025
Viewed by 309
Abstract
Chronic kidney disease (CKD) is associated with the systemic accumulation of uremic toxins (UTs) due to impaired renal elimination. Among these, indoxyl sulfate (IS) and p-cresyl sulfate (PCS) are particularly challenging because of their high protein binding and limited removal by dialysis. In [...] Read more.
Chronic kidney disease (CKD) is associated with the systemic accumulation of uremic toxins (UTs) due to impaired renal elimination. Among these, indoxyl sulfate (IS) and p-cresyl sulfate (PCS) are particularly challenging because of their high protein binding and limited removal by dialysis. In addition to renal excretion, the transport of IS and PCS, and their microbiota-derived precursors, indole and p-cresol, across key physiological barriers—the intestinal barrier, blood–brain barrier, and renal proximal tubule—critically influences their distribution and elimination. This review provides an overview of transporter-mediated mechanisms involved in the disposition of IS, PCS, and their microbial precursors, indole and p-cresol. It also examines how these UTs may interact with commonly prescribed drugs in CKD, particularly those that share transporter pathways as substrates or inhibitors. These drug–toxin interactions may influence the pharmacokinetics and toxicity of IS and PCS, but remain poorly characterized and largely overlooked in clinical settings. A better understanding of these processes may guide future efforts to optimize pharmacotherapy and support more informed management of CKD patients, particularly in the context of polypharmacy. Full article
(This article belongs to the Special Issue Transporters in Health and Disease)
Show Figures

Figure 1

18 pages, 8365 KiB  
Article
Shedding of GPP130 by PC7 and Furin: Potential Implication in Lung Cancer Progression
by Priyanka Prabhala, Stephanie Duval, Alexandra Evagelidis, Maïlys Le Dévéhat, Vatsal Sachan and Nabil G. Seidah
Int. J. Mol. Sci. 2025, 26(13), 6164; https://doi.org/10.3390/ijms26136164 - 26 Jun 2025
Viewed by 299
Abstract
From a previously performed proteomics screen, GPP130, or Golgi phosphoprotein of 130 kDa, was identified as a potential substrate of the proprotein convertase 7 (PC7; PCSK7). GPP130 is a type-II transmembrane protein with a luminal domain containing endosomal and Golgi-retrieval determinants, enabling a [...] Read more.
From a previously performed proteomics screen, GPP130, or Golgi phosphoprotein of 130 kDa, was identified as a potential substrate of the proprotein convertase 7 (PC7; PCSK7). GPP130 is a type-II transmembrane protein with a luminal domain containing endosomal and Golgi-retrieval determinants, enabling a unique trafficking route. Most of the previous work on GPP130 relates to its binding and retrograde trafficking of the Shiga toxin. However, its cellular biology and its biochemical characterization remain understudied. Recently, GPP130 was reported to be implicated in cell cycle progression and cell proliferation in head and neck cancer cells. This led us to analyze the cBioPortal for Cancer Genomics, revealing that the GPP130/GOLIM4 gene is amplified in many cancers, including lung, ovarian, and cervical. This observation led us to use the A549 lung cancer cell line to investigate the growth-regulating roles of endogenous and overexpressed GPP130 and to analyze the impact of its cleavage/shedding by PC7 and/or Furin on cellular growth. Our cell-based assays suggest that GPP130 is a novel pro-protein convertase substrate that increases cell proliferation in A549, SKOV3, and HeLa cells, and that the latter activity is enhanced following its cleavage by PC7 and/or Furin into a membrane-bound N-terminal product and secreted C-terminal fragments. This novel work sheds light on the cell biology of the poorly characterized GPP130, its proliferative activity, and modulation upon its shedding by PC7 and Furin in lung cancer progression. Full article
Show Figures

Figure 1

9 pages, 514 KiB  
Communication
Human Serum Albumin Affinity for Putrescine Using ITC and STD-NMR
by Vida Dehghan Niestanak, Ryan McKay, Marcello Tonelli and Larry D. Unsworth
Int. J. Mol. Sci. 2025, 26(13), 6084; https://doi.org/10.3390/ijms26136084 - 25 Jun 2025
Viewed by 340
Abstract
Understanding the binding interactions between protein-bound uremic toxins (PBUTs) and human serum albumin (HSA) is critical for advancing treatments for chronic kidney disease (CKD). While previous studies have suggested that putrescine, a diamine PBUT, exhibits moderate binding affinity to HSA, this study provides [...] Read more.
Understanding the binding interactions between protein-bound uremic toxins (PBUTs) and human serum albumin (HSA) is critical for advancing treatments for chronic kidney disease (CKD). While previous studies have suggested that putrescine, a diamine PBUT, exhibits moderate binding affinity to HSA, this study provides evidence of the contrary. Using isothermal titration calorimetry and saturation transfer difference nuclear magnetic resonance , we demonstrate that putrescine’s interaction with HSA is weak, non-specific, and thermodynamically negligible in the range of conditions studied. Unlike earlier studies relying on spectroscopy techniques such as UV–visible absorption and fluorescence, which may overestimate binding strength, the results presented here highlight the limitations of indirect methodologies and underscore the importance of more sensitive approaches for accurate energy characterization. Our findings suggest that putrescine only weakly interacts non-specifically with HSA and may bind more preferentially to other plasma proteins, contributing to its accumulation in CKD patients. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

14 pages, 3788 KiB  
Article
Identification of Streptococcus pneumoniae Sortase A Inhibitors and the Interactive Mechanism
by Guizhen Wang, Jiahui Lu, Jingyao Wen, Yifan Duan, Hanbing Zhou, Xinli Peng and Zhandong Li
Crystals 2025, 15(7), 594; https://doi.org/10.3390/cryst15070594 - 24 Jun 2025
Viewed by 329
Abstract
Streptococcus pneumoniae (S. pneumoniae) Sortase A (SrtA) anchors virulence proteins to the surface of the cell wall by recognizing and cleaving the LPXTG motif. These toxins help bacteria adhere to and colonize host cells, promote biofilm formation, and trigger host inflammatory [...] Read more.
Streptococcus pneumoniae (S. pneumoniae) Sortase A (SrtA) anchors virulence proteins to the surface of the cell wall by recognizing and cleaving the LPXTG motif. These toxins help bacteria adhere to and colonize host cells, promote biofilm formation, and trigger host inflammatory responses. Therefore, SrtA is an ideal target for the development of new preparations for S. pneumoniae. In this study, we found that phloretin (pht) and phlorizin (phz) exhibited excellent affinities for SrtA based on virtual screening experiments. We analyzed the interactive mechanism between pht, phz, and alnusone (aln, a reported S. pneumoniae SrtA inhibitor) and SrtA based on molecular dynamics simulation experiments. The results showed that these inhibitors bound to the active pocket of SrtA, and the root mean square deviation (RMSD) and distance analyses showed that these compounds and SrtA maintained stable configuration and binding during the assay. The binding free energy analysis showed that both electrostatic forces (ele), van der Waals forces (vdw), and hydrogen bonds (Hbonds) promoted the binding between pht, phz, and SrtA; however, for the binding of aln and SrtA, the vdw force was much stronger than ele, and Hbonds were not found. The binding free energy decomposition showed that HIS141, ILE143, and PHE119 contributed more energy to promote pht and SrtA binding; ARG215, ASP188, and LEU210 contributed more energy to promote phz and SrtA binding; and HIS141, ASP209, and ARG215 contributed more energy to promote aln and SrtA binding. Finally, the transpeptidase activity of SrtA decreased significantly when treated with different concentrations of pht, phz, or aln, which inhibited S. pneumoniae biofilm formation and adhesion to A549 cells without affecting normal bacterial growth. These results suggest that pht, phtz, and aln are potential materials for the development of novel inhibitors against S. pneumoniae infection. Full article
Show Figures

Figure 1

17 pages, 1733 KiB  
Article
Humanized VHH-hFc Fusion Proteins Targeting the L-HN Fragment of Tetanus Toxin Provided Protection In Vivo
by Yating Li, Kexuan Cheng, Jiazheng Guo, Yujia Jiang, Qinglin Kang, Rong Wang, Peng Du, Chen Gao, Yunzhou Yu, Zhixin Yang, Wei Wang and Jiansheng Lu
Antibodies 2025, 14(2), 48; https://doi.org/10.3390/antib14020048 - 13 Jun 2025
Viewed by 409
Abstract
Background: Tetanus toxin, produced by Clostridium tetani, is the second deadliest known toxin. Antibodies capable of neutralizing tetanus toxin (TeNT) are vital for preventing and treating tetanus disease. Methods: Herein, we screened thirty-six single variable domains on a heavy chain (VHHs) binding [...] Read more.
Background: Tetanus toxin, produced by Clostridium tetani, is the second deadliest known toxin. Antibodies capable of neutralizing tetanus toxin (TeNT) are vital for preventing and treating tetanus disease. Methods: Herein, we screened thirty-six single variable domains on a heavy chain (VHHs) binding to the light chain (L) and the translocation domain (HN) (L-HN) fragment of TeNT from a phage-display library. Then, the L-HN-specific clones were identified, humanized, and fused with a human fragment crystallizable region (hFc) to form humanized VHH-hFc fusion proteins. Results: The humanized VHH-hFc fusion proteins TL-16-h1-hFc, TL-25-h1-hFc, and TL-34-h1-hFc possessed potent efficacy with high binding affinity, specificity, and neutralizing activity. Only 0.3125 μg was required for TL-16-h1-hFc or TL-25-h1-hFc, and 0.625 μg was required for TL-34-h1-hFc to provide full protection against 10 × Lethal Dose 50 (LD50) TeNT. In the prophylactic setting, 125 μg/kg of TL-16-h1-hFc or TL-25-h1-hFc provided full protection even when they were injected 12 days before exposure to 10 × LD50 TeNT, while TL-34-h1-hFc was less effective. In the therapeutic setting, 25 μg/kg of TL-16-h1-hFc or TL-25-h1-hFc could provide complete protection when administered 24 h after exposure to 5 × LD50 TeNT, while TL-34-h1-hFc required 50 μg/kg. Conclusion: Our results suggest that TL-16-h1-hFc, TL-25-h1-hFc, and TL-34-h1-hFc provide a bright future for the development of anti-TeNT preventive or therapeutic drugs. Full article
Show Figures

Figure 1

34 pages, 2009 KiB  
Review
The Multifaceted Role of Calcium Signaling in Regulated Necrosis
by Eric Perez-Rivera, Claudia Plasencia and Uris Ros
Biomolecules 2025, 15(6), 854; https://doi.org/10.3390/biom15060854 - 11 Jun 2025
Viewed by 2237
Abstract
Calcium is a versatile ion that regulates diverse intracellular processes, including cell death and survival, cytokine and chemokine production, lipid scrambling, and immune cell activation. In regulated necrosis, an early increase in cytosolic calcium is a hallmark of pathways such as pyroptosis, necroptosis, [...] Read more.
Calcium is a versatile ion that regulates diverse intracellular processes, including cell death and survival, cytokine and chemokine production, lipid scrambling, and immune cell activation. In regulated necrosis, an early increase in cytosolic calcium is a hallmark of pathways such as pyroptosis, necroptosis, and ferroptosis, and resembles the calcium surge triggered by pore-forming toxins. The complexity of calcium signaling is orchestrated by specialized channels in various cellular compartments and calcium-binding proteins that respond to localized calcium concentrations. However, the coordination of this intricate code during regulated necrosis and its connections to other calcium-driven processes remains poorly understood. This review provides an overview of the molecular mechanisms of calcium signaling in regulated necrosis, analyzing parallels with pore-forming toxin-mediated membrane damage to uncover nodes that are shared by these seemingly independent pathways. We also discuss advanced techniques for studying calcium dynamics, with high precision, that can be applied to study regulated necrosis. Calcium signaling emerges as a central hub where necrotic cell death pathways converge, shaping the unique signatures of dying cells and influencing their communication with the immune system. This integrated perspective highlights the complex and multifaceted role of calcium in cells and its implications for fundamental cellular processes. Full article
(This article belongs to the Special Issue Necrotic Cell Death Mechanisms and Therapies)
Show Figures

Figure 1

20 pages, 335 KiB  
Review
From Physicochemical Classification to Multidimensional Insights: A Comprehensive Review of Uremic Toxin Research
by Mario Cozzolino, Lorenza Magagnoli and Paola Ciceri
Toxins 2025, 17(6), 295; https://doi.org/10.3390/toxins17060295 - 10 Jun 2025
Viewed by 669
Abstract
Chronic kidney disease (CKD) is a global health burden, with uremic toxins (UTs) playing a central role in its pathophysiology. In this review, we systematically examined the evolution of UT classification from the 2003 European Uremic Toxin Work Group (EUTox) system based on [...] Read more.
Chronic kidney disease (CKD) is a global health burden, with uremic toxins (UTs) playing a central role in its pathophysiology. In this review, we systematically examined the evolution of UT classification from the 2003 European Uremic Toxin Work Group (EUTox) system based on molecular weight and protein-binding properties to the 2023 multidimensional framework integrating clinical outcomes, clearance technologies, and artificial intelligence. We highlighted the toxicity mechanisms of UTs across the cardiovascular, immune, and nervous systems and evaluated traditional (e.g., low-/high-flux hemodialysis) and advanced (e.g., high-cutoff dialysis and hemoadsorption) clearance strategies. Despite progress, challenges persist in toxin detection, clearance efficiency, and personalized therapy. Future directions include multi-omics-based biomarker discovery, optimized dialysis membranes, advanced adsorption technology, and AI-driven treatment personalization. This synthesis aims to bridge translational gaps and guide precision medicine in nephrology. Full article
Show Figures

Figure 1

27 pages, 3028 KiB  
Article
Integrated Assessment of Antibacterial Activity, Polyphenol Composition, Molecular Docking, and ADME Properties of Romanian Oak and Fir Honeydew Honeys
by Calin Hulea, Diana Obistioiu, Anca Hulea, Mukhtar Adeiza Suleiman, Doris Floares (Oarga), Ersilia Alexa, Ilinca Merima Imbrea, Alina-Georgeta Neacșu, Marius Pentea, Cosmin Alin Popescu and Florin Imbrea
Antibiotics 2025, 14(6), 592; https://doi.org/10.3390/antibiotics14060592 - 8 Jun 2025
Viewed by 758
Abstract
Background: This study evaluated the polyphenolic composition, antibacterial activity, molecular docking interactions, and pharmacokinetic properties of Romanian oak and fir honeydew honeys. Methods: Spectrophotometric methods quantified total phenolic, flavonoid contents and antioxidant activity, and individual polyphenols were identified via HPLC-MS. Antibacterial efficacy against [...] Read more.
Background: This study evaluated the polyphenolic composition, antibacterial activity, molecular docking interactions, and pharmacokinetic properties of Romanian oak and fir honeydew honeys. Methods: Spectrophotometric methods quantified total phenolic, flavonoid contents and antioxidant activity, and individual polyphenols were identified via HPLC-MS. Antibacterial efficacy against Gram-positive and Gram-negative bacteria was evaluated by determining the bacterial inhibition percentage and minimum inhibitory concentrations. The bioactive compounds identified via LC-MS analysis were used to further delineate the possible antibacterial activities in silico. Molecular docking was carried out to predict the binding interactions and complex formation of the identified compounds against protein crystal structures of the bacteria used in this study. Additionally, the pharmacokinetic profile of compounds with high inhibitory potential was assessed via ADMET (absorption, Distribution, Metabolism, Excretion, toxicity) predictors to ascertain their value. Results: Fir honeydew honey showed higher total phenolic (844.5 mg GAE/kg) and flavonoid contents (489.01 mg QUE/kg) compared to oak honeydew honey, correlating with more potent antioxidant activity (IC50 = 5.16 mg/mL). In vitro antimicrobial tests indicated a stronger inhibitory effect of fir honeydew honey, especially against Gram-positive strains like S. aureus, S. pyogenes, and L. monocytogenes, alongside certain Gram-negative strains such as E. coli and H. influenzae. Oak honeydew honey displayed selective antimicrobial action, particularly against P. aeruginosa and S. typhimurium. The docking outcomes showed rutin, rosmarinic acid, beta resorcylic acid, quercetin, ferulic acid, and p-coumaric acid have high inhibitory activities characterised by binding affinities and binding interactions against shiga toxin, riboflavin synthase, ATP-binding sugar transporter-like protein, undecaprenyl diphosphate synthase, putative lipoprotein, sortase A, and immunity protein, making them key contributors to the honey’s antimicrobial activity. Moreover, beta-resorcylic acid, quercetin, ferulic acid, and p-coumaric acid revealed interesting ADMET scores that qualify honey to serve as a good antimicrobial agent. Conclusions: These findings support their potential use as natural antibacterial agents and emphasise the value of integrating chemical, biological, and computational approaches for multidisciplinary characterisations. Full article
Show Figures

Figure 1

23 pages, 2440 KiB  
Article
LRP1 Shedding in Ricin-Induced Lung Injury: A Cell-Specific Response to Toxin Exposure
by Anita Sapoznikov, Yentl Evgy, Moshe Aftalion and Reut Falach
Int. J. Mol. Sci. 2025, 26(12), 5448; https://doi.org/10.3390/ijms26125448 - 6 Jun 2025
Viewed by 438
Abstract
Ricin is a highly potent toxin that causes severe lung injury upon inhalation by initiating a complex cascade of cellular responses that ultimately leads to cell death. The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor involved in various physiological processes, [...] Read more.
Ricin is a highly potent toxin that causes severe lung injury upon inhalation by initiating a complex cascade of cellular responses that ultimately leads to cell death. The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor involved in various physiological processes, including ricin-mediated toxicity. This study explores the role of LRP1 shedding in the development of ricin-induced lung injury. Analysis of bronchoalveolar lavage fluid (BALF) from ricin-intoxicated mice and swine showed a significant increase in soluble LRP1 (sLRP1) levels, whereas serum LRP1 levels remained largely unchanged, suggesting the lungs are the primary source of sLRP1 release. In vitro assays demonstrated the formation of ricin-sLRP1 complexes, indicating that sLRP1 in BALF retained ricin-binding capability. Flow cytometric analysis of lung cells revealed a reduction in both the percentage and total number of LRP1-expressing cells following ricin exposure. Further investigation of specific lung cell populations showed that alveolar epithelial type II (AT-II) cells, despite experiencing significant injury, exhibited minimal LRP1 shedding. No shedding of LRP1 occurred in neutrophils. In contrast, fibroblasts, which were resistant to ricin-induced cell death, exhibited increased shedding of LRP1 and a corresponding decrease in membrane-bound LRP1 expression. This shedding of the LRP1 ectodomain was mediated by metalloproteinases. Immunohistochemical staining further confirmed decreased LRP1 expression in fibroblasts from ricin-exposed mice. Macrophages also showed substantial LRP1 shedding, despite undergoing significant depletion. These findings highlight the complex cell-specific nature of LRP1 shedding in response to ricin intoxication and suggests the potential role of LRP1 in modulation of cellular susceptibility and resistance to ricin-induced lung injury. Full article
Show Figures

Figure 1

21 pages, 3980 KiB  
Article
Binding Capacity and Adsorption Stability of Uremic Metabolites to Albumin-Modified Magnetic Nanoparticles
by Indu Sharma, Agatha Milley, Lun Zhang, Jiamin Zheng, Ethan Lockwood, David S. Wishart, Marcello Tonelli and Larry D. Unsworth
Int. J. Mol. Sci. 2025, 26(11), 5366; https://doi.org/10.3390/ijms26115366 - 3 Jun 2025
Viewed by 364
Abstract
Kidney disease causes the retention of uremic metabolites in blood, which is associated with many comorbidities. Hemodialysis does not properly clear many metabolites, including large, middle-sized, and small protein-bound uremic toxins (PBUTs). Adsorption strategies for metabolite removal require the development of engineered adsorbents [...] Read more.
Kidney disease causes the retention of uremic metabolites in blood, which is associated with many comorbidities. Hemodialysis does not properly clear many metabolites, including large, middle-sized, and small protein-bound uremic toxins (PBUTs). Adsorption strategies for metabolite removal require the development of engineered adsorbents with tailored surfaces to increase the binding of desired metabolites. Albumin is uniquely positioned for modifying blood-contacting surfaces to absorb uremic metabolites, as it (i) minimizes non-specific protein adsorption and (ii) binds a range of molecules at Sudlow Sites I and II with different affinities. It is unknown if albumin-modified surfaces retain the adsorption qualities of solution-free albumin, namely, adsorption stability or specificity. Herein, albumin was covalently attached to iron oxide nanoparticles and characterized using multiple methods. Metabolite adsorption was conducted by incubating particles in a model solution of thirty-three uremic metabolites associated with kidney failure. Adsorption efficiency, selectivity, and stability were affected by albumin concentration and incubation time. Metabolite adsorption was found to change with time, and it was more effective on albumin-modified particles than unmodified controls. The findings outlined in this paper are crucial for the design of next-generation advanced blood-contacting materials to enhance dialysis and blood purification for patients with kidney disease. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

18 pages, 9688 KiB  
Article
The Role of a Conserved Arg-Asp Pair in the Structure and Function of Tetanus Neurotoxin
by Elizabeth A. Wilson, Ashtyn N. Bevans and Michael R. Baldwin
Toxins 2025, 17(6), 273; https://doi.org/10.3390/toxins17060273 - 30 May 2025
Viewed by 1176
Abstract
Tetanus, a severe and life-threatening illness caused by Clostridium tetani, produces symptoms such as muscle spasms, muscle stiffness and seizures caused by the production of tetanus neurotoxin (TeNT). TeNT causes spastic paralysis through the inhibition of neurotransmission in spinal inhibitory interneurons. This [...] Read more.
Tetanus, a severe and life-threatening illness caused by Clostridium tetani, produces symptoms such as muscle spasms, muscle stiffness and seizures caused by the production of tetanus neurotoxin (TeNT). TeNT causes spastic paralysis through the inhibition of neurotransmission in spinal inhibitory interneurons. This is achieved, in part, through pH-triggered membrane insertion of the translocation (HCT) domain, which delivers the catalytic light-chain (LC) domain to the cytosol. While the function of HCT is well defined, the mechanism by which it accomplishes this task is largely unknown. Based on the crystal structure of tetanus neurotoxin, we identified potential polar interactions between arginine 711, tryptophan 715 and aspartate 821 that appear to be evolutionarily conserved across the clostridial neurotoxin family. We show that the disruption of the Asp-Arg pair in a beltless HCT variant (bHCT) results in changes in thermal stability without significant alterations to the overall secondary structure. ANS (1-anilino-8-napthalene sulfonate) binding studies, in conjunction with liposome permeabilization assays, demonstrate that mutations at R711 or D821 trigger interactions with the membrane at higher pH values compared to wildtype bHCT. Interestingly, we show that the introduction of the D821N mutation into LHNT (LC-HCT only), but not the holotoxin, resulted in the increased cleavage of VAMP 2 in cortical neurons relative to the wildtype protein. This suggests that, as observed for botulinum toxin A, the receptor-binding domain is not necessary for LC translocation but rather helps determine the pH threshold of membrane insertion. The mutation of W715 did not result in detectable changes in the activity of either bHCT or the holotoxin, suggesting that it plays only a minor role in stabilizing the structure of the toxin. We conclude that the protonation of D821 at low pH disrupts interactions with R711 and W715, helping to drive the conformational refolding of HCT needed for membrane insertion and the subsequent translocation of the LC. Full article
(This article belongs to the Section Bacterial Toxins)
Show Figures

Figure 1

Back to TopTop