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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (259)

Search Parameters:
Keywords = glycan receptors

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 1573 KiB  
Article
Polyvalent Mannuronic Acid-Coated Gold Nanoparticles for Probing Multivalent Lectin–Glycan Interaction and Blocking Virus Infection
by Rahman Basaran, Darshita Budhadev, Eleni Dimitriou, Hannah S. Wootton, Gavin J. Miller, Amy Kempf, Inga Nehlmeier, Stefan Pöhlmann, Yuan Guo and Dejian Zhou
Viruses 2025, 17(8), 1066; https://doi.org/10.3390/v17081066 - 30 Jul 2025
Viewed by 286
Abstract
Multivalent lectin–glycan interactions (MLGIs) are vital for viral infection, cell-cell communication and regulation of immune responses. Their structural and biophysical data are thus important, not only for providing insights into their underlying mechanisms but also for designing potent glycoconjugate therapeutics against target MLGIs. [...] Read more.
Multivalent lectin–glycan interactions (MLGIs) are vital for viral infection, cell-cell communication and regulation of immune responses. Their structural and biophysical data are thus important, not only for providing insights into their underlying mechanisms but also for designing potent glycoconjugate therapeutics against target MLGIs. However, such information remains to be limited for some important MLGIs, significantly restricting the research progress. We have recently demonstrated that functional nanoparticles, including ∼4 nm quantum dots and varying sized gold nanoparticles (GNPs), densely glycosylated with various natural mono- and oligo- saccharides, are powerful biophysical probes for MLGIs. Using two important viral receptors, DC-SIGN and DC-SIGNR (together denoted as DC-SIGN/R hereafter), as model multimeric lectins, we have shown that α-mannose and α-manno-α-1,2-biose (abbreviated as Man and DiMan, respectively) coated GNPs not only can provide sensitive measurement of MLGI affinities but also reveal critical structural information (e.g., binding site orientation and mode) which are important for MLGI targeting. In this study, we produced mannuronic acid (ManA) coated GNPs (GNP-ManA) of two different sizes to probe the effect of glycan modification on their MLGI affinity and antiviral property. Using our recently developed GNP fluorescence quenching assay, we find that GNP-ManA binds effectively to both DC-SIGN/R and increasing the size of GNP significantly enhances their MLGI affinity. Consistent with this, increasing the GNP size also significantly enhances their ability to block DC-SIGN/R-augmented virus entry into host cells. Particularly, ManA coated 13 nm GNP potently block Ebola virus glycoprotein-driven entry into DC-SIGN/R-expressing cells with sub-nM levels of EC50. Our findings suggest that GNP-ManA probes can act as a useful tool to quantify the characteristics of MLGIs, where increasing the GNP scaffold size substantially enhances their MLGI affinity and antiviral potency. Full article
(This article belongs to the Special Issue Role of Lectins in Viral Infections and Antiviral Intervention)
Show Figures

Figure 1

21 pages, 3177 KiB  
Review
Galectin-3: Integrator of Signaling via Hexosamine Flux
by Mana Mohan Mukherjee, Devin Biesbrock and John Allan Hanover
Biomolecules 2025, 15(7), 1028; https://doi.org/10.3390/biom15071028 - 16 Jul 2025
Viewed by 296
Abstract
Galectin-3 (Gal-3) is a β-galactoside-binding lectin that mediates diverse signaling events in multiple cell types, including immune cells. It is also a prognostic indicator for multiple clinically important disorders, including cardiovascular disease. Gal-3 binds to cell surface glycans to form lattices that modulate [...] Read more.
Galectin-3 (Gal-3) is a β-galactoside-binding lectin that mediates diverse signaling events in multiple cell types, including immune cells. It is also a prognostic indicator for multiple clinically important disorders, including cardiovascular disease. Gal-3 binds to cell surface glycans to form lattices that modulate surface receptor signaling and internalization. However, the tissue-specific regulation of Gal-3 surface expression remains poorly understood. Here, we review evidence for the involvement of Gal-3 in cell surface signaling, intranuclear events, and intracellular trafficking. Our focus will be on the O-GlcNAc modification as a regulator of Gal-3 biosynthesis, non-canonical secretion, and recycling. We argue that the nutrient-driven cytoplasmic hexosamine biosynthetic pathway (HBP) and endomembrane transport mechanisms generate unique pools of nucleotide sugars. The differing levels of nucleotide sugars in the cytosol, endoplasmic reticulum (ER), and Golgi apparatus generate differential thresholds for the responsiveness of O-GlcNAc cycling, N- and O-linked glycan synthesis/branching, and glycolipid synthesis. By regulating Gal-3 synthesis and non-canonical secretion, O-GlcNAc cycling may serve as a nexus constraining Gal-3 cell surface expression and lattice formation. This homeostatic feedback mechanism would be critical under conditions where extensive glycan synthesis and branching in the endomembrane system and on the cell surface are maintained by elevated hexosamine synthesis. Thus, O-GlcNAc cycling and Gal-3 synergize to regulate Gal-3 secretion and influence cellular signaling. In humans, Gal-3 serves as an early-stage prognostic indicator for heart disease, kidney disease, viral infection, autoimmune disease, and neurodegenerative disorders. Since O-GlcNAc cycling has also been linked to these pathologic states, exploring the interconnections between O-GlcNAc cycling and Gal-3 expression and synthesis is likely to emerge as an exciting area of research. Full article
(This article belongs to the Special Issue Cell Biology and Biomedical Application of Galectins)
Show Figures

Figure 1

17 pages, 695 KiB  
Review
Genetic Diseases of Fucosylation: Insights from Model Organisms
by Muhammad T. Ameen and Curtis R. French
Genes 2025, 16(7), 800; https://doi.org/10.3390/genes16070800 - 3 Jul 2025
Viewed by 927
Abstract
Fucosylation plays a fundamental role in maintaining cellular functions and biological processes across all animals. As a form of glycosylation, it involves the biochemical addition of fucose, a six-carbon monosaccharide, to biological molecules like lipids, proteins, and glycan chains. This modification is essential [...] Read more.
Fucosylation plays a fundamental role in maintaining cellular functions and biological processes across all animals. As a form of glycosylation, it involves the biochemical addition of fucose, a six-carbon monosaccharide, to biological molecules like lipids, proteins, and glycan chains. This modification is essential for optimizing cellular interactions required for receptor-ligand binding, cell adhesion, immune responses, and development. Disruptions in cellular fucose synthesis or in the mechanisms enabling its transfer to other molecules have been linked to human disease. Inherited defects in the fucosylation pathway are rare, with about thirty patients described. Through genome-wide association studies (GWAS), variants in fucosylation pathway genes have been associated with complex diseases such as glaucoma and stroke, and somatic mutations are often found in cancers. Recent studies have applied targeted genetic animal models to elucidate the mechanisms through which disruptions in fucosylation contribute to disease pathogenesis and progression. Key focus areas include GDP-fucose synthesis through de novo or salvage pathways, GDP-fucose transport into the Golgi and endoplasmic reticulum (ER), and its transfer by fucosyltransferases (FUTs) or protein O-fucosyltransferases (POFUTs) onto acceptor molecules. Loss or gain of function fucosylation gene mutations in animal models such as mice, zebrafish, and invertebrates have provided insights into some fucosylation disease pathogenesis. This review aims to bring together these findings, summarizing key insights from existing animal studies to possibly infer fucosylation disease mechanisms in humans. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

13 pages, 2255 KiB  
Article
Solid-Phase Oligosaccharide Synthesis with Highly Complexed Peptidoglycan Fragments
by Yuichiro Kadonaga, Ning Wang, Atsushi Shimoyama, Yukari Fujimoto and Koichi Fukase
Molecules 2025, 30(13), 2787; https://doi.org/10.3390/molecules30132787 - 28 Jun 2025
Viewed by 417
Abstract
Peptidoglycan (PGN) is a component of bacterial cell walls; its fragments are recognized by the cytoplasmic receptors Nod1 and Nod2, thereby promoting the production of inflammatory cytokines and antibodies. To further elucidate these biological defense mechanisms, a large and stable supply of the [...] Read more.
Peptidoglycan (PGN) is a component of bacterial cell walls; its fragments are recognized by the cytoplasmic receptors Nod1 and Nod2, thereby promoting the production of inflammatory cytokines and antibodies. To further elucidate these biological defense mechanisms, a large and stable supply of the PGN fragments via chemical synthesis is essential. However, the synthesis and purification of long PGN fragments are quite challenging due to their low solubility. In this study, we efficiently synthesized PGN fragments via solid-phase oligosaccharide synthesis (SPOS). Using the JandaJel™ Wang resin (JJ-Wang), an octasaccharide glycan chain of PGN was constructed by repeating glycosylation reactions to elongate β-1,4-linked disaccharide units composed of MurNAc and GlcNAc. To enhance reactivity, glycosylation was performed in a mixed solvent comprising C4F9OEt/CH2Cl2/THF with the intention of promoting substrate concentration onto the solid support through the fluorophobic effect, affording the PGN octasaccharide in a 19% overall yield (10 steps). Subsequently, after deprotection of the O-Fmoc, N-Troc, and ethyl ester groups, N- and O-acetylation proceeded smoothly, owing to the high swelling property of JJ-Wang. Peptide condensation with L-Ala-D-isoGln(OBn) and carboxylic acids was also achieved. Finally, cleavage of the PGN fragment from the resin with TFA afforded the desired octasaccharide with dipeptides in a 2.3% overall yield (15 steps). Full article
(This article belongs to the Special Issue 30th Anniversary of Molecules—Recent Advances in Organic Chemistry)
Show Figures

Graphical abstract

35 pages, 2933 KiB  
Review
NEU1-Mediated Extracellular Vesicle Glycosylation in Alzheimer’s Disease: Mechanistic Insights into Intercellular Communication and Therapeutic Targeting
by Mohd Adnan, Arif Jamal Siddiqui, Fevzi Bardakci, Malvi Surti, Riadh Badraoui and Mitesh Patel
Pharmaceuticals 2025, 18(6), 921; https://doi.org/10.3390/ph18060921 - 19 Jun 2025
Viewed by 683
Abstract
Alzheimer’s disease (AD), a progressive neurodegenerative disorder, is marked by the pathological accumulation of amyloid-β plaques and tau neurofibrillary tangles, both of which disrupt neuronal communication and function. Emerging evidence highlights the role of extracellular vesicles (EVs) as key mediators of intercellular communication, [...] Read more.
Alzheimer’s disease (AD), a progressive neurodegenerative disorder, is marked by the pathological accumulation of amyloid-β plaques and tau neurofibrillary tangles, both of which disrupt neuronal communication and function. Emerging evidence highlights the role of extracellular vesicles (EVs) as key mediators of intercellular communication, particularly in the propagation of pathological proteins in AD. Among the regulatory factors influencing EV composition and function, neuraminidase 1 (NEU1), a lysosomal sialidase responsible for desialylating glycoproteins has gained attention for its involvement in EV glycosylation. This review explores the role of NEU1 in modulating EV glycosylation, with particular emphasis on its influence on immune modulation and intracellular trafficking pathways and the subsequent impact on intercellular signaling and neurodegenerative progression. Altered NEU1 activity has been associated with abnormal glycan profiles on EVs, which may facilitate the enhanced spread of amyloid-β and tau proteins across neural networks. By regulating glycosylation, NEU1 influences EV stability, targeting and uptake by recipient cells, primarily through the desialylation of surface glycoproteins and glycolipids, which alters the EV charge, recognition and receptor-mediated interactions. Targeting NEU1 offers a promising therapeutic avenue to restore EV homeostasis and reduces pathological protein dissemination. However, challenges persist in developing selective NEU1 inhibitors and effective delivery methods to the brain. Furthermore, altered EV glycosylation patterns may serve as potential biomarkers for early AD diagnosis and monitoring. Overall, this review highlights the importance of NEU1 in AD pathogenesis and advocates for deeper investigation into its regulatory functions, with the aim of advancing therapeutic strategies and biomarker development for AD and related neurological disabilities. Full article
(This article belongs to the Special Issue Pharmacotherapy for Alzheimer’s Disease)
Show Figures

Graphical abstract

13 pages, 665 KiB  
Review
Galectin-12 in the Regulation of Sebocyte Proliferation, Lipid Metabolism, and Immune Responses
by Ching-Han Tsao, Wei-Chen Hsieh, Feng-Jen Lin, Fu-Tong Liu and Ri-Yao Yang
Biomolecules 2025, 15(6), 837; https://doi.org/10.3390/biom15060837 - 8 Jun 2025
Viewed by 576
Abstract
Galectin-12, a member of the galectin family of glycan-binding proteins, exhibits restricted tissue distribution, primarily in adipocytes and sebocytes. In sebocytes, it modulates the cell cycle, influences lipid metabolism through interactions with peroxisome proliferator-activated receptor γ (PPARγ), and exerts immunomodulatory functions by activating [...] Read more.
Galectin-12, a member of the galectin family of glycan-binding proteins, exhibits restricted tissue distribution, primarily in adipocytes and sebocytes. In sebocytes, it modulates the cell cycle, influences lipid metabolism through interactions with peroxisome proliferator-activated receptor γ (PPARγ), and exerts immunomodulatory functions by activating immune signaling pathways. Dysregulation of sebocyte homeostasis, lipid metabolism, and immune responses contributes to the pathogenesis of a number of skin diseases, underscoring the therapeutic potential of targeting galectin-12. The review summarizes and discusses current developments in the field to foster future research in this important but underexplored galectin. Full article
(This article belongs to the Special Issue Cell Biology and Biomedical Application of Galectins)
Show Figures

Figure 1

17 pages, 2209 KiB  
Article
N-Glycosylation as a Key Requirement for the Positive Interaction of Integrin and uPAR in Glioblastoma
by Gretel Magalí Ferreira, Hector Adrian Cuello, Aylen Camila Nogueira, Jeremias Omar Castillo, Selene Rojo, Cynthia Antonella Gulino, Valeria Inés Segatori and Mariano Rolando Gabri
Int. J. Mol. Sci. 2025, 26(11), 5310; https://doi.org/10.3390/ijms26115310 - 31 May 2025
Viewed by 3171
Abstract
Integrin αV (IαV) and the urokinase-type plasminogen activator receptor (uPAR) are key mediators of tumor malignancy in Glioblastoma. This study aims to characterize IαV/uPAR interaction in GBM and investigate the role played by glycans in this scenario. Protein expression and interaction were confirmed [...] Read more.
Integrin αV (IαV) and the urokinase-type plasminogen activator receptor (uPAR) are key mediators of tumor malignancy in Glioblastoma. This study aims to characterize IαV/uPAR interaction in GBM and investigate the role played by glycans in this scenario. Protein expression and interaction were confirmed via confocal microscopy and co-immunoprecipitation. The role of N-glycosylation was evaluated using Swainsonine (SW) and PNGase F. IαV glycoproteomic analysis was performed by mass spectrometry. Sialic acids and glycan structures in IαV/uPAR interaction were tested using neuraminidase A (NeuA) and lectin interference assays, respectively. Protein expression and their interaction were detected in GBM cells, but not in low-grade glioma cells, even in cells transfected to overexpress uPAR. SW, PNGase, and NeuA treatments significantly reduced IαV/uPAR interaction. Also, lectin interference assays indicated that β1-6 branched glycans play a crucial role in this interaction. Analysis of the IαV glycosylation profile revealed the presence of complex and hybrid N-glycans in GBM, while only oligomannose N-glycans were identified in low-grade glioma. N-glycosylation inhibition and sialic acid removal reduced AKT phosphorylation. Our findings demonstrate, for the first time, the interaction between IαV and uPAR in GBM cells, highlighting the essential role of N-glycosylation, particularly β1-6 branched glycans and sialic acids. Full article
(This article belongs to the Special Issue Glycobiology of Health and Diseases)
Show Figures

Figure 1

13 pages, 9470 KiB  
Article
The Binding of Brazilin from C. sappan to the Full-Length SARS-CoV-2 Spike Proteins
by Phonphiphat Bamrung, Borvornwat Toviwek, Firdaus Samsudin, Phoom Chairatana, Peter John Bond and Prapasiri Pongprayoon
Int. J. Mol. Sci. 2025, 26(9), 4100; https://doi.org/10.3390/ijms26094100 - 25 Apr 2025
Viewed by 564
Abstract
The emergence of coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has become a global issue since 2019. The prominent characteristic of SARS-CoV-2 is the presence of the spike (S) protein protruding from the virus particle envelope. The S protein [...] Read more.
The emergence of coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has become a global issue since 2019. The prominent characteristic of SARS-CoV-2 is the presence of the spike (S) protein protruding from the virus particle envelope. The S protein is a major drug and vaccine target because it initiates the key step in infection. Medicinal herbs are a potential treatment option to enhance immunity to fight viral infections. Caesalpinia sappan L. has been reported to display promising anti-viral activities. Specifically, brazilin (BRA), a major bioactive compound in C. sappan, was reported to play a role in inhibiting viral infection. Thus, the ability of BRA as a COVID-19 treatment was tested. The S protein was used as the BRA target of this work. Understanding the binding mechanism of BRA to the S protein is crucial for future utilisation of C. sappan as a COVID-19 treatment or other coronavirus-caused pandemics. Here, we performed molecular docking of BRA onto the S protein receptor binding domain (RBD) and multimerisation (MM) pockets. Molecular dynamics (MD) simulations were conducted to study the stability of binding to glycosylated and non-glycosylated S protein constructs. BRA can bind to the Receptor-binding motif (RBM) on an RBD surface stably; however, it is too large to fit into the MM pocket, resulting in dissociation. Nonetheless, BRA is bound by residues near the S1/S2 interface. We found that glycosylation has no effect on BRA binding, as the proposed binding site is far from any glycans. Our results thus indicate that C. sappan may act as a promising preventive and therapeutic alternative for COVID-19 treatment. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

22 pages, 2019 KiB  
Article
A Single-Domain VNAR Nanobody Binds with High-Affinity and Selectivity to the Heparin Pentasaccharide Fondaparinux
by Martha Gschwandtner, Rupert Derler, Elisa Talker, Christina Trojacher, Nina Gubensäk, Walter Becker, Tanja Gerlza, Zangger Klaus, Pawel Stocki, Frank S. Walsh, Julia Lynn Rutkowski and Andreas Kungl
Int. J. Mol. Sci. 2025, 26(9), 4045; https://doi.org/10.3390/ijms26094045 - 24 Apr 2025
Viewed by 866
Abstract
Glycosaminoglycans (GAGs) are key ligands for proteins involved in physiological and pathological processes. Specific GAG-binding patterns are rarely identified, with the heparin pentasaccharide as an Antithrombin-III ligand being the best characterized. Generating glycan-specific antibodies is difficult due to their size, pattern dispersion, and [...] Read more.
Glycosaminoglycans (GAGs) are key ligands for proteins involved in physiological and pathological processes. Specific GAG-binding patterns are rarely identified, with the heparin pentasaccharide as an Antithrombin-III ligand being the best characterized. Generating glycan-specific antibodies is difficult due to their size, pattern dispersion, and flexibility. Single-domain variable new antigen receptors (VNAR nanobodies) from nurse sharks are highly soluble, stable, and versatile. Their unique properties suggest advantages over conventional antibodies, particularly for challenging biotherapeutic targets. Here we have used VNAR semi-synthetic phage libraries to select high-affinity fondaparinux-binding VNARs that did not show cross-reactivity with other GAG species. Competition ELISA and surface plasmon resonance identified a single fondaparinux-selective VNAR clone. This VNAR exhibited an extraordinarily stable protein fold: the beta-strands are stabilized by a robust hydrophobic network, as revealed by heteronuclear NMR. Docking fondaparinux to the VNAR structure revealed a large contact surface area between the CDR3 loop of the antibody and the glycan. Fusing the VNAR with a human Fc domain resulted in a stable product with a high affinity for fondaparinux (Kd = 9.3 × 10−8 M) that could efficiently discriminate between fondaparinux and other glycosaminoglycans. This novel glycan-targeting screening technology represents a promising therapeutic strategy for addressing GAG-related diseases. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

20 pages, 2078 KiB  
Review
Bacterial Sialidases: Biological Significance and Application
by Stephan Engibarov, Yana Gocheva, Irina Lazarkevich and Rumyana Eneva
Appl. Biosci. 2025, 4(2), 17; https://doi.org/10.3390/applbiosci4020017 - 1 Apr 2025
Viewed by 1010
Abstract
This review summarizes recent findings on the diverse roles of bacterial sialidases in microbial biology. Bacterial sialidases, also known as neuraminidases, are exog α-lycosidases that cleave terminal sialic acid residues from a number of complex compounds designated as sialoglycoconjugates (glycoproteins, glycolipids and oligosaccharides). [...] Read more.
This review summarizes recent findings on the diverse roles of bacterial sialidases in microbial biology. Bacterial sialidases, also known as neuraminidases, are exog α-lycosidases that cleave terminal sialic acid residues from a number of complex compounds designated as sialoglycoconjugates (glycoproteins, glycolipids and oligosaccharides). Metabolically, they are involved in sialic acid catabolism, providing energy, carbon and nitrogen sources. Catabolic degradation of sialic acids is a physiological feature that can be considered an important virulence factor in pathogenic microorganisms. Sialidases play a pivotal role in host–pathogen interactions and promotion of bacterial colonization. The activity of these enzymes enables bacterial adhesion, biofilm formation, tissue invasion, and also provides immune evasion by exposing cryptic receptors and modifying immune components. Many different perspectives are being developed for the potential application of sialidases. In the field of medicine, they are being explored as appropriate targets for antimicrobials, vaccines, diagnostic preparations and in tumor immunotherapy. In the field of enzymatic synthesis, they are used for the regioselective production of oligosaccharide analogs, enzymatic separation of isoenzymes and as a tool for structural analysis of sialylated glycans, among other applications. Full article
Show Figures

Figure 1

44 pages, 3571 KiB  
Review
Protein O-Fucosyltransferases: Biological Functions and Molecular Mechanisms in Mammals
by Huilin Hao, Benjamin M. Eberand, Mark Larance and Robert S. Haltiwanger
Molecules 2025, 30(7), 1470; https://doi.org/10.3390/molecules30071470 - 26 Mar 2025
Cited by 1 | Viewed by 2032
Abstract
Domain-specific O-fucosylation is an unusual type of glycosylation, where the fucose is directly attached to the serine or threonine residues in specific protein domains via an O-linkage. O-fucosylated proteins play critical roles in a wide variety of biological events and [...] Read more.
Domain-specific O-fucosylation is an unusual type of glycosylation, where the fucose is directly attached to the serine or threonine residues in specific protein domains via an O-linkage. O-fucosylated proteins play critical roles in a wide variety of biological events and hold important therapeutic values, with the most studied being the Notch receptors and ADAMTS proteins. O-fucose glycans modulate the function of the proteins they modify and are closely associated with various diseases including cancer. In mammals, alongside the well-documented protein O-fucosyltransferase (POFUT) 1-mediated O-fucosylation of epidermal growth factor-like (EGF) repeats and POFUT2-mediated O-fucosylation of thrombospondin type 1 repeats (TSRs), a new type of O-fucosylation was recently identified on elastin microfibril interface (EMI) domains, mediated by POFUT3 and POFUT4 (formerly FUT10 and FUT11). In this review, we present an overview of our current knowledge of O-fucosylation, integrating the latest findings and with a particular focus on its biological functions and molecular mechanisms. Full article
(This article belongs to the Section Chemical Biology)
Show Figures

Figure 1

20 pages, 12053 KiB  
Article
Integrated mRNA and miRNA Omics Analyses Reveal Transcriptional Regulation of the Tolerance Traits by Aquatica leii in Response to High Temperature
by Chao Liu, Jiapeng Li, Lihong Yan, Yuting Zhu, Zikun Li, Chengquan Cao and Yiping Wang
Insects 2025, 16(3), 316; https://doi.org/10.3390/insects16030316 - 18 Mar 2025
Viewed by 620
Abstract
Within the context of global warming, understanding the molecular mechanisms behind physiological plasticity and local adaptation is essential for insect populations. This study performed an integrated miRNA and mRNA analysis on Aquatica leii larvae exposed to temperatures of 20 °C, 24 °C, 28 [...] Read more.
Within the context of global warming, understanding the molecular mechanisms behind physiological plasticity and local adaptation is essential for insect populations. This study performed an integrated miRNA and mRNA analysis on Aquatica leii larvae exposed to temperatures of 20 °C, 24 °C, 28 °C, and 32 °C. Under varying thermal conditions, 1983 genes exhibited differential expression (i.e., DEGs). These genes showed significant enrichment in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to carbohydrate metabolism, glycan biosynthesis and metabolism. Notably, we detected that the “neuroactive ligand–receptor interaction” signaling pathway, which is involved in environmental information processing, was significantly upregulated in the 28 °C and 32 °C treatment groups. This indicates that starting at 28 °C, A. leii needs to maintain normal cellular physiological functions by regulating ligand–receptor binding and signal transduction. Furthermore, 220 differentially expressed miRNAs (DEMs) were detected under the different temperature treatment conditions. An interaction network was constructed between key DEMs and DEGs, revealing 12 significant DEM-DEG regulatory pairs in A. leii under different temperature treatments. We found three miRNA-mRNA candidate modules that could be involved in A. leii’s response to high temperature, including ggo-miR-1260b and ptr-miR-1260b/RN001_010114, CM069438.1_43851/RN001_014852, and CM069438.1_43851/RN001_014877. Our data provide deeper insights into the molecular responses of A. leii to the high temperature at the miRNA and mRNA levels. Full article
(This article belongs to the Special Issue Aquatic Insects: Ecology, Diversity and Conservation)
Show Figures

Figure 1

16 pages, 2258 KiB  
Article
Glycosylated Receptor-Binding-Domain-Targeting Mucosal Vaccines Protect Against SARS-CoV-2 Omicron and MERS-CoV
by Xiaoqing Guan, Abhishek K. Verma, Qian Liu, Melissa Palacios, Abby E. Odle, Stanley Perlman and Lanying Du
Vaccines 2025, 13(3), 293; https://doi.org/10.3390/vaccines13030293 - 10 Mar 2025
Viewed by 1002
Abstract
Background. The pathogenic coronaviruses (CoVs) MERS-CoV and SARS-CoV-2, which are responsible for the MERS outbreak and the COVID-19 pandemic, respectively, continue to infect humans, with significant adverse outcomes. There is a continuing need to develop mucosal vaccines against these respiratory viral pathogens to [...] Read more.
Background. The pathogenic coronaviruses (CoVs) MERS-CoV and SARS-CoV-2, which are responsible for the MERS outbreak and the COVID-19 pandemic, respectively, continue to infect humans, with significant adverse outcomes. There is a continuing need to develop mucosal vaccines against these respiratory viral pathogens to prevent entry and replication at mucosal sites. The receptor-binding domain (RBD) of the CoV spike (S) protein is a critical vaccine target, and glycan masking is a unique approach for designing subunit vaccines with improved neutralizing activity. Methods. We evaluated the efficacy of mucosal immunity, broad neutralizing activity, and cross-protection afforded by a combined glycosylated mucosal subunit vaccine encoding the RBDs of the original SARS-CoV-2 strain (SARS2-WT-RBD), the Omicron-XBB.1.5 variant (SARS2-Omi-RBD), and MERS-CoV (MERS-RBD). Results. Intranasal administration of the three-RBD protein cocktail induced effective, durable IgA and systemic IgG antibodies specific for the S protein of these CoVs, thereby neutralizing infection by pseudotyped SARS-CoV-2-WT, Omicron-XBB.1.5, and MERS-CoV. The mucosal vaccine cocktail protected immunized mice from challenge with SARS-CoV-2 Omicron-XBB.1.5 and MERS-CoV, leading to a significant reduction in the viral titers in the lungs. By contrast, the individual glycosylated RBD proteins only induced such immune responses and neutralizing antibodies against either SARS-CoV-2 or MERS-CoV, protecting against subsequent challenge with either SARS-CoV-2 or MERS-CoV; they did not provide simultaneous protection against both CoVs. Conclusions. This study describes a unique strategy for designing efficacious mucosal subunit vaccines that induce durable mucosal immunity, cross-neutralizing activity, and cross-protection against SARS-CoV-2 and MERS-CoV, highlighting the potential for the design of mucosal vaccines against other pathogens. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Figure 1

15 pages, 4154 KiB  
Article
Bioinformatic Selection of Mannose-Specific Lectins from Allium genus as SARS-CoV-2 Inhibitors Analysing Protein–Protein Interaction
by Stefan Isaković, Milan Senćanski, Vladimir Perović, Kristina Stevanović and Ivana Prodić
Life 2025, 15(2), 162; https://doi.org/10.3390/life15020162 - 23 Jan 2025
Cited by 1 | Viewed by 1176
Abstract
Mannose-specific lectins are carbohydrate-binding proteins known for their antiviral potential. This study uses a bioinformatic approach to investigate the possibility of lectins from Allium sativum (garlic) and Allium ursinum (wild garlic) as inhibitors of SARS-CoV-2 entry. The information spectrum method (ISM) identified key [...] Read more.
Mannose-specific lectins are carbohydrate-binding proteins known for their antiviral potential. This study uses a bioinformatic approach to investigate the possibility of lectins from Allium sativum (garlic) and Allium ursinum (wild garlic) as inhibitors of SARS-CoV-2 entry. The information spectrum method (ISM) identified key interaction frequencies between the SARS-CoV-2 spike protein and these lectins, explicitly targeting the receptor-binding domain (RBD) and glycosylated asparagine residues, including N234. Lectins from Allium species showed a high affinity for oligomannose-type glycans on the spike protein, potentially blocking virus entry by preventing the spike-ACE2 receptor interaction. We propose that Allium lectins are promising candidates for further experimental validation as SARS-CoV-2 inhibitors, offering potential therapeutic applications in managing viral infections. Full article
(This article belongs to the Section Biochemistry, Biophysics and Computational Biology)
Show Figures

Figure 1

19 pages, 1386 KiB  
Review
Galectins and Liver Diseases
by Shima Mimura, Asahiro Morishita, Kyoko Oura, Kei Takuma, Mai Nakahara, Tomoko Tadokoro, Koji Fujita, Joji Tani and Hideki Kobara
Int. J. Mol. Sci. 2025, 26(2), 790; https://doi.org/10.3390/ijms26020790 - 18 Jan 2025
Cited by 2 | Viewed by 1234
Abstract
Galectins are widely distributed throughout the animal kingdom, from marine sponges to mammals. Galectins are a family of soluble lectins that specifically recognize β-galactoside-containing glycans and are categorized into three subgroups based on the number and function of their carbohydrate recognition domains (CRDs). [...] Read more.
Galectins are widely distributed throughout the animal kingdom, from marine sponges to mammals. Galectins are a family of soluble lectins that specifically recognize β-galactoside-containing glycans and are categorized into three subgroups based on the number and function of their carbohydrate recognition domains (CRDs). The interaction of galectins with specific ligands mediates a wide range of biological activities, depending on the cell type, tissue context, expression levels of individual galectin, and receptor involvement. Galectins affect various immune cell processes through both intracellular and extracellular mechanisms and play roles in processes, such as apoptosis, angiogenesis, and fibrosis. Their importance has increased in recent years because they are recognized as biomarkers, therapeutic agents, and drug targets, with many other applications in conditions such as cardiovascular diseases and cancer. However, little is known about the involvement of galectins in liver diseases. Here, we review the functions of various galectins and evaluate their roles in liver diseases. Full article
(This article belongs to the Special Issue Galectins (Gals))
Show Figures

Figure 1

Back to TopTop