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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

Search Results (314)

Search Parameters:
Keywords = microdomain

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 11000 KB  
Hypothesis
Serotonergic Signaling Rewired: A Lipid Raft-Controlled Model of Synaptic Transmission Grounded in the Fundamental Parameters of Biological Systems
by Jacques Fantini, Marine Lefebvre, Nouara Yahi and Henri Chahinian
Life 2026, 16(1), 118; https://doi.org/10.3390/life16010118 - 13 Jan 2026
Viewed by 307
Abstract
Serotonergic signaling is traditionally conceived as a transient, vesicle-mediated process restricted to the synaptic cleft. Here, we propose an expanded model in which serotonin can also be inserted into the plasma membrane of neurons and glial cells, forming a stable, membrane-associated reservoir that [...] Read more.
Serotonergic signaling is traditionally conceived as a transient, vesicle-mediated process restricted to the synaptic cleft. Here, we propose an expanded model in which serotonin can also be inserted into the plasma membrane of neurons and glial cells, forming a stable, membrane-associated reservoir that prolongs its availability beyond classical synaptic timescales. In this framework, the synapse emerges not as a simple neurotransmitter–receptor interface but as a dynamic, multiscale medium where membrane order, hydration, and quantum-level processes jointly govern information flow. Two temporal “tunnels” appear to regulate serotonin bioavailability: its aggregation in synaptic vesicles during exocytosis, and its cholesterol-dependent insertion into neuronal and glial membranes at the tripartite synapse. Lipid raft microdomains enriched in cholesterol and gangliosides thus act as active regulators of a continuum between transient and constitutive serotonin signaling. This extended serotonergic persistence prompts a reconsideration of current pharmacological models and the action of antidepressants such as fluoxetine, which not only inhibits the serotonin transporter (SERT) but also accumulates in lipid rafts, perturbs raft organization, and alters serotonin–cholesterol equilibria, contributing to SERT-independent effects. Grounded in the recently established fundamental parameters of biological systems, this model invites a broader, quantum-informed rethinking of synaptic transmission. Full article
(This article belongs to the Section Medical Research)
Show Figures

Figure 1

18 pages, 3817 KB  
Article
Selective Budding of SARS-CoV-Like Particles from Glycolipid-Enriched Membrane Lipid Rafts and Host Gene Modulation
by Manoj K. Pastey, Yue Huang and Barney Graham
Microorganisms 2026, 14(1), 159; https://doi.org/10.3390/microorganisms14010159 - 10 Jan 2026
Viewed by 210
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) assembles and buds from the Golgi apparatus or the ER membrane, but the specific membrane microdomains utilized during this process remain underexplored. Here, we show that co-expression of the SARS-CoV structural proteins S, M, and N in [...] Read more.
Severe acute respiratory syndrome coronavirus (SARS-CoV) assembles and buds from the Golgi apparatus or the ER membrane, but the specific membrane microdomains utilized during this process remain underexplored. Here, we show that co-expression of the SARS-CoV structural proteins S, M, and N in HEK-293T cells is sufficient to generate genome-free SARS-CoV-like virus-like particles (VLPs), which preferentially bud from glycolipid-enriched membrane lipid raft microdomains. Immunofluorescence microscopy using raft-selective dyes (DiIC16) and spike-specific antibodies revealed strong co-localization of VLPs with lipid rafts. Detergent-resistant membrane analysis and sucrose gradient centrifugation further confirmed the presence of S protein in buoyant, raft-associated fractions alongside the raft marker CD44. Importantly, pharmacological disruption of rafts with methyl-β-cyclodextrin reduced VLP budding and S protein partitioning into raft domains, underscoring the requirement for intact lipid rafts in assembly. Additionally, our data support lipid raft-associated proteins’ (e.g., FNRA, VIM, CD59, RHOA) roles in modulating cellular responses conducive to viral replication and assembly. These findings highlight lipid rafts as crucial platforms for SARS-CoV morphogenesis and suggest new avenues for vaccine and antiviral development using VLPs and raft-targeting therapeutics. Full article
(This article belongs to the Special Issue Coronavirus: Epidemiology, Diagnosis, Pathogenesis and Control)
Show Figures

Figure 1

16 pages, 7964 KB  
Article
Metallic Flexible NiTi Wire Microcrack Transducer for Label-Free Impedimetric Sensing of Escherichia coli
by Gizem Özlü Türk and Mehmet Çağrı Soylu
Biosensors 2026, 16(1), 54; https://doi.org/10.3390/bios16010054 - 10 Jan 2026
Viewed by 235
Abstract
Flexible biosensors offer rapid and low-cost diagnostics but are often limited by the mechanical and electrochemical instability of polymer-based designs in biological media. Here, we introduce a metallic flexible microcrack transducer that exploits the intrinsic deformability of superelastic nickel–titanium (NiTi) for label-free impedimetric [...] Read more.
Flexible biosensors offer rapid and low-cost diagnostics but are often limited by the mechanical and electrochemical instability of polymer-based designs in biological media. Here, we introduce a metallic flexible microcrack transducer that exploits the intrinsic deformability of superelastic nickel–titanium (NiTi) for label-free impedimetric detection. Mechanical bending of NiTi wires spontaneously generates martensitic-phase microcracks whose metal–gap–metal geometry forms the active transduction sites, where functional interfacial layers and captured analytes modulate the local dielectric environment and govern the impedance response. Our approach imparts a novel dielectric character to the alloy, enabling its unexplored application in the megahertz (MHz) frequency domain (0.01–10 MHz) where native NiTi is merely conductive. Functionalization with Escherichia coli (E. coli)-specific antibodies renders these microdomains biologically active. This effectively transforms the mechanically induced microcracks into tunable impedance elements driven by analyte binding. The γ-bent NiTi sensors achieved stable and quantitative detection of E. coli ATCC 25922 in sterile human urine, with a detection limit of 64 colony forming units (CFU) mL−1 within 45 min, without redox mediators, external labels, or amplification steps. This work pioneers the use of martensitic microcrack networks, mimicking self-healing behavior in a superelastic alloy as functional transduction elements, defining a new class of metallic flexible biosensors that integrate mechanical robustness, analytical reliability, and scalability for point-of-care biosensing. Full article
(This article belongs to the Special Issue Functional Materials for Biosensing Applications (2nd Edition))
Show Figures

Graphical abstract

35 pages, 3152 KB  
Review
AI-Resolved Protein Energy Landscapes, Electrodynamics, and Fluidic Microcircuits as a Unified Framework for Predicting Neurodegeneration
by Cosmin Pantu, Alexandru Breazu, Stefan Oprea, Matei Serban, Razvan-Adrian Covache-Busuioc, Octavian Munteanu, Nicolaie Dobrin, Daniel Costea and Lucian Eva
Int. J. Mol. Sci. 2026, 27(2), 676; https://doi.org/10.3390/ijms27020676 - 9 Jan 2026
Viewed by 209
Abstract
Research shows that neurodegenerative processes do not develop from a single “broken” biochemistry process; rather, they develop when a complex multi-physics environment gradually loses its ability to stabilize the neuron via a collective action between the protein, ion, field and fluid dynamics of [...] Read more.
Research shows that neurodegenerative processes do not develop from a single “broken” biochemistry process; rather, they develop when a complex multi-physics environment gradually loses its ability to stabilize the neuron via a collective action between the protein, ion, field and fluid dynamics of the neuron. The use of new technologies such as quantum-informed molecular simulation (QIMS), dielectric nanoscale mapping, fluid dynamics of the cell, and imaging of perivascular flow are allowing researchers to understand how the collective interactions among proteins, membranes and their electrical properties, along with fluid dynamics within the cell, form a highly interconnected dynamic system. These systems require fine control over the energetic, mechanical and electrical interactions that maintain their coherence. When there is even a small change in the protein conformations, the electric properties of the membrane, or the viscosity of the cell’s interior, it can cause changes in the high dimensional space in which the system operates to lose some of its stabilizing curvature and become prone to instability well before structural pathologies become apparent. AI has allowed researchers to create digital twin models using combined physical data from multiple scales and to predict the trajectory of the neural system toward instability by identifying signs of early deformation. Preliminary studies suggest that deviations in the ergodicity of metabolic–mechanical systems, contraction of dissipative bandwidth, and fragmentation of attractor basins could be indicators of vulnerability. This study will attempt to combine all of the current research into a cohesive view of the role of progressive loss of multi-physics coherence in neurodegenerative disease. Through integration of protein energetics, electrodynamic drift, and hydrodynamic irregularities, as well as predictive modeling utilizing AI, the authors will provide mechanistic insights and discuss potential approaches to early detection, targeted stabilization, and precision-guided interventions based on neurophysics. Full article
Show Figures

Figure 1

33 pages, 1141 KB  
Review
The Protonic Brain: Nanoscale pH Dynamics, Proton Wires, and Acid–Base Information Coding in Neural Tissue
by Valentin Titus Grigorean, Catalina-Ioana Tataru, Cosmin Pantu, Felix-Mircea Brehar, Octavian Munteanu and George Pariza
Int. J. Mol. Sci. 2026, 27(2), 560; https://doi.org/10.3390/ijms27020560 - 6 Jan 2026
Viewed by 249
Abstract
Emerging research indicates that neuronal activity is maintained by an architectural system of protons in a multi-scale fashion. Proton architecture is formed when organelles (such as mitochondria, endoplasmic reticulum, lysosomes, synaptic vesicles, etc.) are coupled together to produce dynamic energy domains. Techniques have [...] Read more.
Emerging research indicates that neuronal activity is maintained by an architectural system of protons in a multi-scale fashion. Proton architecture is formed when organelles (such as mitochondria, endoplasmic reticulum, lysosomes, synaptic vesicles, etc.) are coupled together to produce dynamic energy domains. Techniques have been developed to visualize protons in neurons; recent advances include near-atomic structural imaging of organelle interfaces using cryo-tomography and nanoscale resolution imaging of organelle interfaces and proton tracking using ultra-fast spectroscopy. Results of these studies indicate that protons in neurons do not diffuse randomly throughout the neuron but instead exist in organized geometric configurations. The cristae of mitochondrial cells create oscillating proton micro-domains that are influenced by the curvature of the cristae, hydrogen bonding between molecules, and localized changes in dielectric properties that result in time-patterned proton signals that can be used to determine the metabolic load of the cell and the redox state of its mitochondria. These proton patterns also communicate to the rest of the cell via hydrated aligned proton-conductive pathways at the mitochon-dria-endoplasmic reticulum junctions, through acidic lipid regions, and through nano-tethered contact sites between mitochondria and other organelles, which are typically spaced approximately 10–25 nm apart. Other proton architectures exist in lysosomes, endosomes, and synaptic vesicles. In each of these organelles, the V-ATPase generates steep concentration gradients across their membranes, controlling the rate of cargo removal from the lumen of the organelle, recycling receptors from the surface of the membrane, and loading neurotransmitters into the vesicles. Recent super-resolution pH mapping has indicated that populations of synaptic vesicles contain significant heterogeneity in the amount of protons they contain, thereby influencing the amount of neurotransmitter released per vesicle, the probability of vesicle release, and the degree of post-synaptic receptor protonation. Additionally, proton gradients in each organelle interact with the cytoskeleton: the protonation status of actin and microtubules influences filament stiffness, protein–protein interactions, and organelle movement, resulting in the formation of localized spatial structures that may possess some type of computational significance. At multiple scales, it appears that neurons integrate the proton micro-domains with mechanical tension fields, dielectric nanodomains, and phase-state transitions to form distributed computing elements whose behavior is determined by the integration of energy flow, organelle geometry, and the organization of soft materials. Alterations to the proton landscape in neurons (e.g., due to alterations in cristae structure, drift in luminal pH, disruption in the hydration-structure of the cell, or imbalance in the protonation of cytoskeletal components) could disrupt the intracellular signaling network well before the onset of measurable electrical or biochemical pathologies. This article will summarize evidence indicating that proton–organelle interaction provides a previously unknown source of energetic substrate for neural computation. Using an integrated approach combining nanoscale proton energy, organelle interface geometry, cytoskeletal mechanics, and AI-based multiscale models, this article outlines current principles and unresolved questions related to the subject area as well as possible new approaches to early detection and precise intervention of pathological conditions related to altered intracellular energy flow. Full article
(This article belongs to the Special Issue Molecular Synapse: Diversity, Function and Signaling)
Show Figures

Figure 1

15 pages, 1720 KB  
Article
Family-Wide Dysregulation of Phosphodiesterases Alters cAMP/cGMP Microdomains in Thoracic Aortic Aneurysm
by Dimitrios E. Magouliotis, Serge Sicouri, Vasiliki Androutsopoulou, Massimo Baudo, Francesco Cabrucci, Prokopis-Andreas Zotos, Andrew Xanthopoulos and Basel Ramlawi
J. Cardiovasc. Dev. Dis. 2026, 13(1), 23; https://doi.org/10.3390/jcdd13010023 - 1 Jan 2026
Viewed by 323
Abstract
Background: Thoracic aortic aneurysm (TAA) is driven by complex molecular mechanisms beyond size thresholds, yet the role of cyclic nucleotide metabolism remains unclear. Phosphodiesterases (PDEs), which hydrolyze cAMP and cGMP in compartmentalized microdomains, act as key regulators of vascular integrity and remodeling. Methods: [...] Read more.
Background: Thoracic aortic aneurysm (TAA) is driven by complex molecular mechanisms beyond size thresholds, yet the role of cyclic nucleotide metabolism remains unclear. Phosphodiesterases (PDEs), which hydrolyze cAMP and cGMP in compartmentalized microdomains, act as key regulators of vascular integrity and remodeling. Methods: We performed a hypothesis-driven, transcriptomic analysis of 20 PDE isoforms using the GSE26155 dataset (43 TAA vs. 43 controls). Raw microarray data underwent background correction, log2 transformation, and false-discovery adjustment. Differential expression, logistic regression, receiver-operating characteristic (ROC) curves, calibration testing, correlation analysis, and interactome/enrichment mapping were conducted. Results: Thirteen PDE isoforms were significantly dysregulated in TAA. Upregulated transcripts included PDE10A, PDE2A, PDE4B, PDE7A, and PDE8A, whereas PDE1A/B/C, PDE3B, PDE5A, PDE6C, and PDE8B were downregulated. PDE10A achieved excellent discrimination for TAA (AUC = 0.838), while other isoforms demonstrated fair discriminatory ability. Correlation architecture revealed coordinated regulation between PDE subfamilies, including inverse relationships between PDE2A and PDE8B (r = −0.68). Interactome analysis highlighted dense connections with cyclic nucleotide and purinergic signaling hubs, enriched in vascular tone, NO–cGMP–PKG, and junctional assembly pathways. Integrating these findings with epigenetic and junctional frameworks suggests that PDE dysregulation promotes endothelial barrier fragility and maladaptive smooth-muscle remodeling. Conclusions: Family-wide PDE dysregulation characterizes human TAA, with PDE10A emerging as a central transcriptomic signature. Altered cAMP/cGMP microdomain signaling aligns with junctional failure and epigenetic control, supporting the potential of PDE isoforms as biomarkers and therapeutic targets. These results provide experimental evidence that cyclic nucleotide hydrolysis is re-wired in TAA, supporting PDE10A as a novel biomarker and therapeutic target that bridges molecular dysregulation with clinical risk stratification in thoracic aortic disease. Full article
Show Figures

Graphical abstract

23 pages, 3017 KB  
Review
Targeting the Cellular Prion Protein as a Biomarker for Stem Cells, Cancer, and Regeneration
by Niccolò Candelise, Nicola Salvatore Orefice, Elisabetta Mantuano and Stefano Martellucci
Biologics 2026, 6(1), 1; https://doi.org/10.3390/biologics6010001 - 24 Dec 2025
Viewed by 466
Abstract
The cellular prion protein (PrPC) displays a functional repertoire that extends well beyond its classical link to transmissible spongiform encephalopathies. Abundant in the nervous system and localized within lipid raft microdomains, PrPC has emerged as a multifunctional signaling platform that [...] Read more.
The cellular prion protein (PrPC) displays a functional repertoire that extends well beyond its classical link to transmissible spongiform encephalopathies. Abundant in the nervous system and localized within lipid raft microdomains, PrPC has emerged as a multifunctional signaling platform that regulates cell differentiation, neurogenesis, neuroprotection, and synaptic plasticity. Recent evidence highlights its dynamic expression in stem cell populations, where it participates in multimolecular complexes that control lineage commitment, particularly during neuronal differentiation. PrPC expression tightly correlates with stem cell status, making it a promising biomarker of stemness and developmental progression. Through interactions with growth factors, extracellular matrix components, and synaptic proteins, PrPC functions as a molecular integrator of signals essential for tissue repair and regeneration. Preclinical studies demonstrate that recombinant PrPC can stimulate neurogenesis and tissue repair, while monoclonal antibodies modulate its physiological and pathological functions. Likewise, cell-based therapies leveraging PrPC-enriched stem cells or PrPC-dependent signaling profiles have shown promise in models of neurodegeneration and ischemia. Conversely, dysregulated PrPC expression has also been observed in solid tumors, where it contributes to cancer cell survival, proliferation, metastasis, and therapy resistance, reinforcing its role as a regulator of cell fate and an oncological target. This review integrates stem cell biology, tissue regeneration, and oncology into a unified framework, offering a novel perspective in which PrPC emerges as a shared molecular hub governing both physiological repair and pathological tumor behavior, opening previously unrecognized conceptual and translational opportunities. Full article
(This article belongs to the Section Protein Therapeutics)
Show Figures

Figure 1

31 pages, 3313 KB  
Article
Inhibition of Sterol Biosynthesis Alters Tubulin Association with Detergent-Insoluble Membranes and Affects Microtubule Organization in Pollen Tubes of Nicotiana tabacum L.
by Elisabetta Onelli, Lilly Maneta-Peyret, Patrick Moreau, Nadia Stroppa, Valeria Berno, Eugenia Cammarota, Marco Caccianiga, Luca Gianfranceschi and Alessandra Moscatelli
Plants 2025, 14(24), 3845; https://doi.org/10.3390/plants14243845 - 17 Dec 2025
Viewed by 358
Abstract
Pollen tube growth entails complex molecular interactions between the cytoskeletal apparatus and membrane trafficking. Tip growth involves polarized distribution of proteins and lipids along the plasma membrane, including liquid-ordered microdomains, rich in sterols and sphingolipids (lipid rafts), in the apical/subapical region of tobacco [...] Read more.
Pollen tube growth entails complex molecular interactions between the cytoskeletal apparatus and membrane trafficking. Tip growth involves polarized distribution of proteins and lipids along the plasma membrane, including liquid-ordered microdomains, rich in sterols and sphingolipids (lipid rafts), in the apical/subapical region of tobacco pollen tubes. Intriguingly, biochemical characterization of detergent-insoluble membranes purified from tobacco pollen tubes revealed the presence of both actin and tubulin. Here, we report that inhibition of sterol biosynthesis altered lipid rafts and lowered the association of tubulin with detergent-insoluble membranes. Our results showed that sterol depletion increased the number of microtubules in the subapical region, altered microtubule distribution and affected microtubule bundling activity. Oryzalin washout experiments also suggested that lipid-ordered domains could play a role in regulating microtubule nucleation/regrowth. Full article
(This article belongs to the Section Plant Cell Biology)
Show Figures

Figure 1

14 pages, 1982 KB  
Article
Repositioning Imipramine for Antiparasitic Effects Against Giardia lamblia
by Xareni Zinereth Herrera-Valero, Sendar Daniel Nery-Flores, Filiberto Gutiérrez-Gutiérrez, Lizeth Guadalupe Campos-Múzquiz, Sandra Cecilia Esparza-González, Raúl Rodríguez-Herrera and Lissethe Palomo-Ligas
Drugs Drug Candidates 2025, 4(4), 56; https://doi.org/10.3390/ddc4040056 - 16 Dec 2025
Viewed by 384
Abstract
Background/Objectives: Giardia lamblia is an intestinal protozoan responsible for giardiasis, a globally prevalent parasitic disease. Current therapeutic options, including nitroimidazoles and benzimidazoles, have increasing treatment failures due to resistance, adverse reactions, and patient non-compliance. Drug repositioning offers a cost-effective strategy for identifying [...] Read more.
Background/Objectives: Giardia lamblia is an intestinal protozoan responsible for giardiasis, a globally prevalent parasitic disease. Current therapeutic options, including nitroimidazoles and benzimidazoles, have increasing treatment failures due to resistance, adverse reactions, and patient non-compliance. Drug repositioning offers a cost-effective strategy for identifying new antigiardial agents. This study aimed to evaluate the in vitro antiparasitic effects and possible mechanisms of action of the tricyclic antidepressant imipramine against G. lamblia trophozoites. Methods: Trophozoites were exposed to increasing concentrations of imipramine (25–125 µM). Growth inhibition and adhesion capacity were quantified using cell counts. Apoptosis- or necrosis-like death was evaluated through Annexin V/PI staining. The expression and distribution of α-tubulin and lipid rafts were analyzed by immunofluorescence microscopy. Finally, the effect of the drug on encystment efficiency was assessed in vitro. Results: Imipramine inhibited G. lamblia trophozoite growth in a concentration-dependent manner, with an IC50 of 42.31 µM at 48 h. The drug significantly reduced adhesion capacity (>90% at 125 µM) and induced apoptosis-like cell death, as evidenced by Annexin V positivity. Immunofluorescence revealed disruption of α-tubulin distribution and lipid raft organization, accompanied by morphological rounding. Moreover, encystment efficiency decreased in a concentration-dependent mode, suggesting interference in the differentiation process. Conclusions: This investigation describes, for the first time, the antigiardial potential of imipramine, which alters cytoskeletal organization, membrane microdomains, and differentiation pathways, ultimately leading to apoptosis-like cell death. These findings position this compound as a promising lead structure and support further exploration of tricyclic antidepressants as scaffolds for the development and optimization of new antiparasitic agents, as well as future studies on their molecular targets and in vivo efficacy. Full article
(This article belongs to the Collection Anti-Parasite Drug Discovery)
Show Figures

Graphical abstract

25 pages, 1719 KB  
Review
Astrocyte-Mediated Plasticity: Multi-Scale Mechanisms Linking Synaptic Dynamics to Learning and Memory
by Masaya Yamamoto and Tetsuya Takano
Cells 2025, 14(24), 1936; https://doi.org/10.3390/cells14241936 - 5 Dec 2025
Viewed by 1977
Abstract
Astrocytes play a pivotal role in shaping synaptic function and in learning, memory, and emotion. Recent studies show that perisynaptic astrocytic processes form structured interactions with pre- and postsynaptic elements, which extends synaptic diversity beyond neuron–neuron connections. Accumulating evidence indicates that astrocytic Ca [...] Read more.
Astrocytes play a pivotal role in shaping synaptic function and in learning, memory, and emotion. Recent studies show that perisynaptic astrocytic processes form structured interactions with pre- and postsynaptic elements, which extends synaptic diversity beyond neuron–neuron connections. Accumulating evidence indicates that astrocytic Ca2+ signaling, gliotransmission, and local translation modulate synaptic efficacy and contribute to the formation and stabilization of memory traces. It is therefore essential to define how astrocytic microdomains, multisynaptic leaflet domains, and network-level ensembles cooperate to regulate circuit computation across space and time. Advances in super-resolution and volumetric in vivo imaging and spatial transcriptomics now enable detailed, cell-type- and compartment-specific analysis of astrocyte–synapse interactions in vivo. In this review, we highlight these approaches and synthesize classical and emerging mechanisms by which astrocytes read neuronal activity, write to synapses, and coordinate network states. We also discuss theoretical frameworks such as neuron–astrocyte associative memory models that formalize astrocytic calcium states as distributed substrates for storage and control. This integrated view provides new insight into the multicellular logic of memory and suggests paths toward understanding and treating neurological and psychiatric disorders. Full article
(This article belongs to the Special Issue Synaptic Plasticity and the Neurobiology of Learning and Memory)
Show Figures

Figure 1

20 pages, 1777 KB  
Review
Regulatory Mechanisms of Lipid Rafts in Remodeling the Tumor Immune Microenvironment of Colorectal Cancer and Targeted Therapeutic Strategies
by Zhihong Cheng, Jian Gu, Yaoyao Lu, Mingdong Cai, Tao Zhang and Jiliang Wang
Biomolecules 2025, 15(12), 1675; https://doi.org/10.3390/biom15121675 - 1 Dec 2025
Viewed by 913
Abstract
Immunotherapy has demonstrated significant efficacy in colorectal cancer (CRC), but its therapeutic effects remain limited in microsatellite stable (MSS) patients, indicating the critical role of the tumor immune microenvironment (TIME) in regulating immune responses. Lipid rafts, dynamic membrane microdomains enriched in cholesterol and [...] Read more.
Immunotherapy has demonstrated significant efficacy in colorectal cancer (CRC), but its therapeutic effects remain limited in microsatellite stable (MSS) patients, indicating the critical role of the tumor immune microenvironment (TIME) in regulating immune responses. Lipid rafts, dynamic membrane microdomains enriched in cholesterol and sphingolipids, have emerged as potential targets for TIME remodeling through their integration of immune signal transduction, enrichment of cell death receptors, and regulation of immune cell functionality. This review outlines the pivotal mediating roles of lipid rafts in cellular survival, death, and tumor progression. Specifically, MSS-type CRC exhibits lipid raft structural remodeling driven by dysregulated lipid metabolism, which fosters multiple immune escape mechanisms through exosome-mediated immunosuppressive signaling, promotion of tumor-associated macrophage (TAM) M2 polarization, enhanced infiltration of regulatory T cells (Tregs), and functional exhaustion of effector cells, such as CD8+ T cells and NK cells. Finally, we discuss targeted therapeutic strategies based on lipid raft characteristics and CRC molecular profiles, proposing an innovative multidimensional treatment framework combining immune checkpoint inhibitors with lipid raft-targeted interventions and chemoradiotherapy. This approach provides theoretical and strategic support for overcoming CRC immunotherapy resistance and advancing clinical translation. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

35 pages, 1987 KB  
Review
The Fluidic Connectome in Brain Disease: Integrating Aquaporin-4 Polarity with Multisystem Pathways in Neurodegeneration
by Felix-Mircea Brehar, Daniel Costea, Calin Petru Tataru, Mugurel Petrinel Rădoi, Alexandru Vlad Ciurea, Octavian Munteanu and Adrian Tulin
Int. J. Mol. Sci. 2025, 26(23), 11536; https://doi.org/10.3390/ijms262311536 - 28 Nov 2025
Viewed by 1610
Abstract
The way in which Aquaporin-4 (AQP4) is localized on the astrocytes’ surface—i.e., with AQP4 channels predominantly located on the endfeet of astrocytes near the blood vessels—represents an important structural element for maintaining brain fluid homeostasis. In addition to this structural function, AQP4 polarity [...] Read more.
The way in which Aquaporin-4 (AQP4) is localized on the astrocytes’ surface—i.e., with AQP4 channels predominantly located on the endfeet of astrocytes near the blood vessels—represents an important structural element for maintaining brain fluid homeostasis. In addition to this structural function, AQP4 polarity also facilitates glymphatic transport, the maintenance of the blood–brain barrier (BBB) functions, ion buffering, and neurotransmitter removal, and helps regulate neurovascular communications. The growing body of literature suggests that the loss of AQP4 polarity—a loss in the organization of AQP4 channels to the perivascular membrane—is associated with increased vascular, inflammatory, and metabolic disturbances in the context of many neurological diseases. As a result, this review attempts to synthesize both experimental and clinical studies to highlight that AQP4 depolarization often occurs in conjunction with early signs of neurodegeneration and neuroinflammation; however, we are aware that the loss of AQP4 polarity is only one factor in a complex pathophysiological environment. This review examines the molecular structure responsible for maintaining the polarity of AQP4—such as dystrophin–syntrophin complexes, orthogonal particle arrays, lipid microdomains, trafficking pathways, and transcriptional regulators—and describes how the vulnerability of these systems to various types of vascular stress, inflammatory signals, energy deficits, and mechanical injury can lead to a loss of AQP4 polarity. Furthermore, we will explore how a loss of AQP4 polarity can lead to the disruption of perivascular fluid movement, changes in blood–brain barrier morphology, enhanced neuroimmune activity, changes in ionic and metabolic balance, and disruptions in the global neural network synchronization. Importantly, we recognize that each of these disruptions will likely occur in concert with other disease-specific mechanisms. Alterations in AQP4 polarity have been observed in a variety of neurological disorders including Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, traumatic brain injury, and glioma; however, we also observe that the same alterations in fluid regulation occur across all of these different diseases, but that no single upstream event accounts for the alteration in polarity. Ultimately, we will outline emerging therapeutic avenues to restore perivascular fluid transport, and will include molecular-based therapeutic agents designed to modify the anchoring of AQP4, methods designed to modulate the state of astrocytes, biomaterials-based drug delivery systems, and therapeutic methods that leverage dynamic modulation of the neurovascular interface. Future advances in multi-omic profiling, spatial proteomics, glymphatic imaging, and artificial intelligence will allow for earlier identification of AQP4 polarity disturbances and potentially allow for the development of more personalized treatment plans. Ultimately, by linking these concepts together, this review aims to frame AQP4 polarity as a modifiable aspect of the “fluidic connectome”, and highlight its importance in maintaining overall brain health across disease states. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Regulation in Blood-Brain Barrier)
Show Figures

Figure 1

19 pages, 880 KB  
Review
Lipid–Protein Interplay in the Regulation of Receptor Tyrosine Kinases
by Mattia Domenichini, Anna Gogna, Camilla Maggi, Elisa Moreschi, Anna Ventura, Martina Codibue, Elisabetta Grillo, Michela Corsini and Stefania Mitola
Cells 2025, 14(23), 1836; https://doi.org/10.3390/cells14231836 - 21 Nov 2025
Viewed by 1042
Abstract
Receptor tyrosine kinases (RTKs), a class of membrane proteins involved in several physiological processes such as growth, survival, angiogenesis, and differentiation, are profoundly influenced by the microenvironment, particularly by surrounding lipids. Lipids coordinate RTK life cycle at multiple steps. First, receptor lipidation is [...] Read more.
Receptor tyrosine kinases (RTKs), a class of membrane proteins involved in several physiological processes such as growth, survival, angiogenesis, and differentiation, are profoundly influenced by the microenvironment, particularly by surrounding lipids. Lipids coordinate RTK life cycle at multiple steps. First, receptor lipidation is a key post-translational modification for receptor-targeting localization. Then, RTK dimerization and activation are regulated by membrane-enriched lipids like phosphatidylserine and phosphoinositides, gangliosides, and Cholesterol, which directly engage RTK juxtamembrane domain or cytoplasmic tail. Eventually, lipids spatially organize RTK signaling within Cholesterol- and sphingolipid-enriched membrane microdomains. These membrane rafts act as dynamic “signalosomes” coordinating receptor clustering, endocytosis, and recycling. Perturbations in lipid composition remodel raft architecture and alter RTK behavior, contributing to pathological conditions such as cancer, metabolic, and neurodegenerative disorders. Emerging lipid-targeted therapies offer a promising way to enhance RTK-directed therapies. This review aims to explore how specific lipid species and membrane domains modulate RTK activation, clustering, and endocytic recycling. By bridging biochemical and pathological perspectives, we discuss how membrane lipid composition reshapes RTK signaling in physiology and pathology, pointing to emerging opportunities for lipid-focused therapeutic modulation. Full article
Show Figures

Graphical abstract

22 pages, 2526 KB  
Article
Untargeted Metabolomics Identifies Faecal Filtrate-Derived Metabolites That Disrupt Clostridioides difficile Metabolism and Confer Gut Barrier Cytoprotection
by Fatimah I. Qassadi, Charlotte Johnson, Karen Robinson, Ruth Griffin, Christos Polytarchou, Dina Kao, Dong-Hyun Kim, Rian L. Griffiths, Zheying Zhu and Tanya M. Monaghan
Int. J. Mol. Sci. 2025, 26(22), 11221; https://doi.org/10.3390/ijms262211221 - 20 Nov 2025
Viewed by 1071
Abstract
Recurrent Clostridioides difficile infection (rCDI) remains a major therapeutic challenge. Although faecal microbiota transplantation (FMT) is highly effective and thought to restore microbial composition and metabolic function, the mechanisms underlying its success are not fully understood. In particular, the contribution of non-bacterial components [...] Read more.
Recurrent Clostridioides difficile infection (rCDI) remains a major therapeutic challenge. Although faecal microbiota transplantation (FMT) is highly effective and thought to restore microbial composition and metabolic function, the mechanisms underlying its success are not fully understood. In particular, the contribution of non-bacterial components such as soluble metabolites remains unclear. Therefore, further investigation is needed to identify the mechanistic drivers of FMT efficacy and clarify how non-bacterial factors contribute to therapeutic outcomes. Here, we applied untargeted three-dimensional Orbitrap secondary ion mass spectrometry (3D OrbiSIMS) to profile faecal metabolic reprogramming in rCDI patients pre- and post-FMT, alongside C. difficile cultures exposed to sterile faecal filtrates. FMT induced extensive metabolic shifts, restoring glyoxylate/dicarboxylate and glycerophosphoinositol pathways and normalising disrupted bile acid and amino acid profiles. Faecal filtrate exposure caused strain-specific metabolic disruption in C. difficile, depleting proline, fumarate and succinate while enriching tryptophan. While multiple metabolite classes were profiled, the most significant functional changes were observed in lipids. Lipidomics identified >3.8-fold enrichment of phosphatidylinositol (PI) species, which localised to bacterial membranes and conferred cytoprotection against C. difficile toxins and other epithelial insults. Spatial metabolomics imaging revealed, for the first time, metabolite compartmentalisation within C. difficile, with proline and succinate broadly distributed across the cell surface and fumarate confined to distinct microdomains, highlighting functional heterogeneity in pathogen metabolism. Collectively, these findings demonstrate that soluble metabolites within faecal filtrates mediate pathogen suppression and epithelial barrier protection, establishing metabolite-driven mechanisms underlying FMT efficacy and identifying PI lipids as candidate post-biotic therapeutics for rCDI. Full article
(This article belongs to the Special Issue Interplay Between the Human Microbiome and Diseases)
Show Figures

Figure 1

10 pages, 1697 KB  
Article
Persistent PbI2-Passivated Microdomains in As-Prepared MAPbI3 Perovskite Thin Film Revealed by Spatially Resolved Photoluminescence and Raman Maps
by Bong-Geun Kim, Jiwon Choi, Taeji Seo, Hyun Sung Kim, Minseok Kwak, Joonkyung Jang, Songyi Lee and Myeongkee Park
Crystals 2025, 15(11), 991; https://doi.org/10.3390/cryst15110991 - 17 Nov 2025
Viewed by 685
Abstract
Methylammonium lead iodide (MAPbI3) perovskite has been widely studied for its optoelectronic properties, but its polycrystalline thin films inevitably contain grain boundaries and defects that degrade performance and stability. PbI2 is often considered a destabilizing agent in perovskites, yet it [...] Read more.
Methylammonium lead iodide (MAPbI3) perovskite has been widely studied for its optoelectronic properties, but its polycrystalline thin films inevitably contain grain boundaries and defects that degrade performance and stability. PbI2 is often considered a destabilizing agent in perovskites, yet it has also been reported to act as a passivation agent, making its role a subject of debate. Here, we performed integrated optical, photoluminescence (PL), and ultra-low-frequency Raman mapping on a 100 μm2 region of MAPbI3 thin films to study the roles of PbI2. The analyses resolved α-phase MAPbI3-rich, PbI2-rich, and mixed-phase domains, revealing heterogeneous PbI2 distribution with PL quenching at defect sites. In addition, after light-induced degradation, PL changes were governed by both the local microstructure and PbI2 distribution. Notably, PbI2-rich regions retained PL, evidencing a protective passivation effect. These findings demonstrate that the beneficial role of PbI2 is key to designing more stable perovskite devices. Full article
(This article belongs to the Section Hybrid and Composite Crystalline Materials)
Show Figures

Figure 1

Back to TopTop