Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96.697
Filter
1.
Biomaterials ; 312: 122707, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121729

ABSTRACT

Polypyrimidine tract-binding protein 1 (PTBP1) regulates numerous alternative splicing events during tumor progression and neurogenesis. Previously, PTBP1 downregulation was reported to convert astrocytes into functional neurons; however, how PTBP1 regulates astrocytic physiology remains unclear. In this study, we revealed that PTBP1 modulated glutamate uptake via ATP1a2, a member of Na+/K+-ATPases, and glutamate transporters in astrocytes. Ptbp1 knockdown altered mitochondrial function and energy metabolism, which involved PTBP1 regulating mitochondrial redox homeostasis via the succinate dehydrogenase (SDH)/Nrf2 pathway. The malfunction of glutamate transporters following Ptbp1 knockdown resulted in enhanced excitatory synaptic transmission in the cortex. Notably, we developed a biomimetic cationic triblock polypeptide system, i.e., polyethylene glycol44-polylysine30-polyleucine10 (PEG44-PLL30-PLLeu10) with astrocytic membrane coating to deliver Ptbp1 siRNA in vitro and in vivo, which approach allowed Ptbp1 siRNA to efficiently cross the blood-brain barrier and target astrocytes in the brain. Collectively, our findings suggest a framework whereby PTBP1 serves as a modulator in glutamate transport machinery, and indicate that biomimetic methodology is a promising route for in vivo siRNA delivery.


Subject(s)
Astrocytes , Glutamic Acid , Heterogeneous-Nuclear Ribonucleoproteins , Homeostasis , NF-E2-Related Factor 2 , Polypyrimidine Tract-Binding Protein , RNA, Small Interfering , Animals , Astrocytes/metabolism , Glutamic Acid/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , NF-E2-Related Factor 2/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mice , Signal Transduction , Cell Membrane/metabolism , Mice, Inbred C57BL , Male , Humans , Mitochondria/metabolism
2.
Methods Mol Biol ; 2852: 105-122, 2025.
Article in English | MEDLINE | ID: mdl-39235739

ABSTRACT

In food industry, Listeria monocytogenes contamination can occur accidentally despite the quality control of raw materials and factory. Decontamination processes or inhibitory effects of ingredients/additives in food products are set up to ensure compliance with hygiene and microbiological criteria. These actions represent stresses for the pathogenic agent, causing fluctuations in its physiological states. Moreover, during these environmental stresses, Listeria monocytogenes can enter in a viable but nonculturable (VBNC) state which is not detected by plate counting but by flow cytometry. This technique coupled with cell staining by fluorescent dyes offers the possibility to assess different physiological states based on different cellular parameters: enzymatic activity, transmembrane integrity, membrane potential, and respiratory activity. In this chapter, we present a method to assess the viability of foodborne pathogens using a double-staining principle based on the assessment of membrane integrity and intracellular esterase activity.


Subject(s)
Flow Cytometry , Listeria monocytogenes , Microbial Viability , Listeria monocytogenes/growth & development , Listeria monocytogenes/physiology , Flow Cytometry/methods , Food Microbiology/methods , Fluorescent Dyes/chemistry , Staining and Labeling/methods , Cell Membrane/metabolism
3.
Methods Mol Biol ; 2852: 135-141, 2025.
Article in English | MEDLINE | ID: mdl-39235741

ABSTRACT

When submitted to environmental stresses, bacteria can modulate its fatty acid composition of membrane phospholipids in order to optimize membrane fluidity. Characterization of bacterial membrane fatty acid profiles is thus an interesting indicator of cellular physiological state. The methodology described here aims to improve the recovering of biofilm cells for the characterization of their fatty acid profiles. The saponification reagent is directly applied on the whole biofilm before the removal of cells from the inert surface. In this way, maximum of the cells and their fatty acids can be recovered from the deepest layers of the biofilm.


Subject(s)
Biofilms , Cell Membrane , Fatty Acids , Biofilms/growth & development , Fatty Acids/metabolism , Cell Membrane/metabolism , Bacteria/metabolism , Phospholipids/metabolism , Membrane Fluidity
4.
Open Biol ; 14(9): 240067, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39288811

ABSTRACT

Calmodulin (CaM) is a ubiquitous calcium-sensitive messenger in eukaryotic cells. It was previously shown that CaM possesses an affinity for diverse lipid moieties, including those found on CaM-binding proteins. These facts, together with our observation that CaM accumulates in membrane-rich protrusions of HeLa cells upon increased cytosolic calcium, motivated us to perform a systematic search for unmediated CaM interactions with model lipid membranes mimicking the cytosolic leaflet of plasma membranes. A range of experimental techniques and molecular dynamics simulations prove unambiguously that CaM interacts with lipid bilayers in the presence of calcium ions. The lipids phosphatidylserine (PS) and phosphatidylethanolamine (PE) hold the key to CaM-membrane interactions. Calcium induces an essential conformational rearrangement of CaM, but calcium binding to the headgroup of PS also neutralizes the membrane negative surface charge. More intriguingly, PE plays a dual role-it not only forms hydrogen bonds with CaM, but also destabilizes the lipid bilayer increasing the exposure of hydrophobic acyl chains to the interacting proteins. Our findings suggest that upon increased intracellular calcium concentration, CaM and the cytosolic leaflet of cellular membranes can be functionally connected.


Subject(s)
Calcium , Calmodulin , Cell Membrane , Cytosol , Lipid Bilayers , Molecular Dynamics Simulation , Phosphatidylserines , Protein Binding , Calmodulin/metabolism , Calmodulin/chemistry , Cell Membrane/metabolism , Calcium/metabolism , Humans , Lipid Bilayers/metabolism , Phosphatidylserines/metabolism , Cytosol/metabolism , Phosphatidylethanolamines/metabolism , HeLa Cells
5.
Sci Rep ; 14(1): 21819, 2024 09 18.
Article in English | MEDLINE | ID: mdl-39294204

ABSTRACT

Toxoplasma gondii is a polarized cell concentrating several secretory organelles at the apical pole. The secretory micronemes come in two sub-populations differentiated by dependence on Rab5A/C in their biogenesis. Calcium-dependent exocytosis of micronemes occurs at the very apical tip and is critical for parasite egress from its host cell, adhesion and invasion of the next cell. Ferlins represent a protein family with roles in exocytosis containing multiple Ca2+-sensing C2 domains. We determined that T. gondii's ferlin 1 (FER1) localized dynamically to the parasite's secretory pathway. FER1 function was dissected by dominant negative overexpression strategies. We demonstrated that FER1 traffics microneme organelles along the following trajectories: (1) Along the cortex to the apical end; (2) To the apical tip for fusion with the plasma membrane; (3) Differential microneme sub-population traffic, and that FER1 could putatively be responsible for microneme protein trafficking. (4) From the trans-Golgi-endosomal network to the subpellicular cortex; (5) Retrograde transport allowing microneme recycling from mother to daughter. Finally, FER1 overexpression triggers a microneme exocytosis burst, supporting the notion that the radially organized micronemes at the apical tip comprise a readily-releasable microneme pool. In summary, FER1 is pivotal for dynamic microneme trafficking, acts differently on the two microneme subpopulations, and acts on the plasma membrane fusion step during microneme exocytosis.


Subject(s)
Exocytosis , Protein Transport , Protozoan Proteins , Toxoplasma , Toxoplasma/metabolism , Protozoan Proteins/metabolism , Organelles/metabolism , Humans , Cell Membrane/metabolism
6.
J Nanobiotechnology ; 22(1): 578, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300463

ABSTRACT

Rheumatoid arthritis (RA) is a debilitating autoimmune disease characterized by chronic joint inflammation and cartilage damage. Current therapeutic strategies often result in side effects, necessitating the development of targeted and safer treatment options. This study introduces a novel nanotherapeutic system, 2-APB@DGP-MM, which utilizes macrophage membrane (MM)-encapsulated nanoparticles (NPs) for the targeted delivery of 2-Aminoethyl diphenylborinate (2-APB) to inflamed joints more effectively. The NPs are designed with a matrix metalloproteinase (MMP)-cleavable peptide, allowing for MMP-responsive drug release within RA microenvironment. Comprehensive in vitro and in vivo assays confirmed the successful synthesis and loading of 2-APB into the DSPE-GPLGVRGC-PEG (DGP) NPs, as well as their ability to repolarize macrophages from a pro-inflammatory M1 to an anti-inflammatory M2 phenotype. The NPs demonstrated high biocompatibility, low cytotoxicity, and enhanced cellular uptake. In a collagen-induced arthritis (CIA) mouse model, intra-articular injection of 2-APB@DGP-MM significantly reduced synovial inflammation and cartilage destruction. Histological analysis corroborated these findings, demonstrating marked improvements in joint structure and delayed disease progression. Above all, the 2-APB@DGP-MM nanotherapeutic system offers a promising and safe approach for RA treatment by modulating macrophage polarization and delivering effective agents to inflamed joints.


Subject(s)
Arthritis, Rheumatoid , Macrophages , Nanoparticles , Animals , Mice , Macrophages/drug effects , Macrophages/metabolism , Arthritis, Rheumatoid/drug therapy , Nanoparticles/chemistry , RAW 264.7 Cells , Male , Mice, Inbred DBA , Arthritis, Experimental/drug therapy , Boron Compounds/chemistry , Boron Compounds/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Humans , Cell Membrane/metabolism , Cell Membrane/drug effects
7.
Cell Death Dis ; 15(9): 675, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39277583

ABSTRACT

Rap2b, a proto-oncogene upregulated in colorectal cancer (CRC), undergoes protein S-palmitoylation at specific C-terminus sites (C176/C177). These palmitoylation sites are crucial for Rap2b localization on the plasma membrane (PM), as mutation of C176 or C177 results in cytosolic relocation of Rap2b. Our study demonstrates that Rap2b influences cell migration and invasion in CRC cells, independent of proliferation, and this activity relies on its palmitoylation. We identify ABHD17a as the depalmitoylating enzyme for Rap2b, altering PM localization and inhibiting cell migration and invasion. EGFR/PI3K signaling regulates Rap2b palmitoylation, with PI3K phosphorylating ABHD17a to modulate its activity. These findings highlight the potential of targeting Rap2b palmitoylation as an intervention strategy. Blocking the C176/C177 sites using an interacting peptide attenuates Rap2b palmitoylation, disrupting PM localization, and suppressing CRC metastasis. This study offers insights into therapeutic approaches targeting Rap2b palmitoylation for the treatment of metastatic CRC, presenting opportunities to improve patient outcomes.


Subject(s)
Cell Membrane , Colorectal Neoplasms , Lipoylation , rap GTP-Binding Proteins , Animals , Humans , Mice , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , ErbB Receptors/metabolism , Mice, Nude , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Mas , rap GTP-Binding Proteins/metabolism , rap GTP-Binding Proteins/genetics , Signal Transduction
8.
J Cell Biol ; 223(12)2024 Dec 02.
Article in English | MEDLINE | ID: mdl-39302311

ABSTRACT

As a consequence of hypoosmotic shock, yeast cells swell rapidly and increase the surface area by ∼20% in 20 s. Approximately, 35% of this surface increase is mediated by the ER-plasma membrane contact sites, specifically the tricalbins, which are required for the delivery of both lipids and the GPI-anchored protein Crh2 from the cortical ER to the plasma membrane. Therefore, we propose a new function for the tricalbins: mediating the fusion of the ER to the plasma membrane at contact sites. This proposed fusion is triggered by calcium influx via the stretch-gated channel Cch1 and is supported by the anoctamin Ist2.


Subject(s)
Cell Membrane , Endoplasmic Reticulum , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Endoplasmic Reticulum/metabolism , Cell Membrane/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Calcium Channels/metabolism , Membrane Lipids/metabolism , Membrane Fusion , Calcium/metabolism , Membrane Proteins/metabolism , Osmotic Pressure
9.
PLoS One ; 19(9): e0308500, 2024.
Article in English | MEDLINE | ID: mdl-39269939

ABSTRACT

Glutathione transferase P1 (GSTP1) is a multi-functional protein that protects cells from electrophiles by catalyzing their conjugation with glutathione, and contributes to the regulation of cell proliferation, apoptosis, and signalling. GSTP1, usually described as a cytosolic enzyme, can localize to other cell compartments and we have reported its strong association with the plasma membrane. In the current study, the hypothesis that GSTP1 is palmitoylated and this modification facilitates its dynamic localization and function was investigated. Palmitoylation is the reversible post-translational addition of a 16-C saturated fatty acid to proteins, most commonly on Cys residues through a thioester bond. GSTP1 in MCF7 cells was modified by palmitate, however, GSTP1 Cys to Ser mutants (individual and Cys-less) retained palmitoylation. Treatment of palmitoylated GSTP1 with 0.1 N NaOH, which cleaves ester bonds, did not remove palmitate. Purified GSTP1 was spontaneously palmitoylated in vitro and peptide sequencing revealed that Cys48 and Cys102 undergo S-palmitoylation, while Lys103 undergoes the rare N-palmitoylation. N-palmitoylation occurs via a stable NaOH-resistant amide bond. Analysis of subcellular fractions of MCF7-GSTP1 cells and a modified proximity ligation assay revealed that palmitoylated GSTP1 was present not only in the membrane fraction but also in the cytosol. GSTP1 isolated from E. coli, and MCF7 cells (grown under fatty acid free or regular conditions), associated with plasma membrane-enriched fractions and this association was not altered by palmitoyl CoA. Overall, GSTP1 is modified by palmitate, at multiple sites, including at least one non-Cys residue. These modifications could contribute to regulating the diverse functions of GSTP1.


Subject(s)
Glutathione S-Transferase pi , Lipoylation , Palmitates , Humans , Glutathione S-Transferase pi/metabolism , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/chemistry , MCF-7 Cells , Palmitates/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Cysteine/metabolism , Protein Processing, Post-Translational , Palmitic Acid/metabolism
10.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273101

ABSTRACT

Bacteriocins are a class of proteins produced by bacteria that are toxic to other bacteria. These bacteriocins play a role in bacterial competition by helping to inhibit potential competitors. In this study, we isolated and purified a novel bacteriocin Pkmh, different from the previously reported bacteriocin PA166, from Pseudomonas sp. strain 166 by ammonium sulfate precipitation, dialysis membrane method, ion exchange chromatography, and gel filtration chromatography. SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) revealed that the molecular weight of Pkmh is approximately 35 kDa. Pkmh exhibited potent antimicrobial activity against bovine Mannheimia haemolytica (M. haemolytica) with low cytotoxicity, and lower hemolytic activity was observed. In addition, Pkmh retained antimicrobial activity at different pH ranges (2-10) and temperature conditions (40, 60, 80, 100 °C). Our analysis of its antimicrobial mechanism showed that Pkmh acts on bacterial cell membranes, increasing their permeability and leading to cell membrane rupture and death. In conclusion, Pkmh exhibited low hemolytic activity, low toxicity, and potent antibacterial effects, suggesting its potential as a promising candidate for clinical therapeutic drugs.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Bacteriocins/pharmacology , Bacteriocins/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Hemolysis/drug effects , Mannheimia haemolytica/drug effects , Pseudomonas/drug effects , Cattle , Microbial Sensitivity Tests , Humans , Molecular Weight , Cell Membrane/drug effects , Cell Membrane/metabolism , Temperature , Hydrogen-Ion Concentration
11.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273619

ABSTRACT

Human lactoferrin (hLf) is an innate host defense protein that inhibits microbial H+-ATPases. This protein includes an ancestral structural motif (i.e., γ-core motif) intimately associated with the antimicrobial activity of many natural Cys-rich peptides. Peptides containing a complete γ-core motif from hLf or other phylogenetically diverse antimicrobial peptides (i.e., afnA, SolyC, PA1b, PvD1, thanatin) showed microbicidal activity with similar features to those previously reported for hLf and defensins. Common mechanistic characteristics included (1) cell death independent of plasma membrane (PM) lysis, (2) loss of intracellular K+ (mediated by Tok1p K+ channels in yeast), (3) inhibition of microbicidal activity by high extracellular K+, (4) influence of cellular respiration on microbicidal activity, (5) involvement of mitochondrial ATP synthase in yeast cell death processes, and (6) increment of intracellular ATP. Similar features were also observed with the BM2 peptide, a fungal PM H+-ATPase inhibitor. Collectively, these findings suggest host defense peptides containing a homologous γ-core motif inhibit PM H+-ATPases. Based on this discovery, we propose that the γ-core motif is an archetypal effector involved in the inhibition of PM H+-ATPases across kingdoms of life and contributes to the in vitro microbicidal activity of Cys-rich antimicrobial peptides.


Subject(s)
Amino Acid Motifs , Proton-Translocating ATPases , Humans , Proton-Translocating ATPases/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Lactoferrin/pharmacology , Lactoferrin/chemistry , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Cysteine/metabolism , Cysteine/chemistry , Candida albicans/drug effects , Cell Membrane/metabolism , Cell Membrane/drug effects
12.
Int J Mol Sci ; 25(17)2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39273636

ABSTRACT

The amyloid cascade hypothesis postulates that extracellular deposits of amyloid ß (Aß) are the primary and initial cause leading to the full development of Alzheimer's disease (AD) with intracellular neurofibrillary tangles; however, the details of this mechanism have not been fully described until now. Our preliminary data, coming from our day-to-day neuropathology practice, show that the primary location of the hyperphosphorylated tau protein is in the vicinity of the cell membrane of dystrophic neurites. This observation inspired us to formulate a hypothesis that presumes an interaction between low-density lipoprotein receptor-related protein 1 (LRP1) and fibrillar aggregates of, particularly, Aß42 anchored at the periphery of neuritic plaques, making internalization of the LRP1-Aß42 complex infeasible and, thus, causing membrane dysfunction, leading to the tauopathy characterized by intracellular accumulation and hyperphosphorylation of the tau protein. Understanding AD as a membrane dysfunction tauopathy may draw attention to new treatment approaches not only targeting Aß42 production but also, perhaps paradoxically, preventing the formation of LRP1-Aß42.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Low Density Lipoprotein Receptor-Related Protein-1 , Tauopathies , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/etiology , Cell Membrane/metabolism , Phosphorylation , Animals , Peptide Fragments/metabolism
13.
Plant Physiol Biochem ; 215: 109078, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39226762

ABSTRACT

-Action potential (AP) of excitable plant cells is an important signaling event that can differentially alter physicochemical and physiological processes in various parts of the same cell. In giant cells of characean algae, the AP propagation has minor effect on photosynthetic electron transport in areas with high activity of plasmalemmal H+-pump but inhibits linear electron flow in regions featuring high passive H+/OH- conductance of the plasma membrane (PM). Uneven spatial distributions of local periplasmic and cytoplasmic pH facilitate the operation of distinct (CO2-dependent and O2-mediated) pathways of photoinduced electron flow, which presumably accounts for differential influence of AP on photosynthesis. The excitation of Chara australis cell in the presence of methyl viologen (MV), a redox mediator with the prooxidant action, provides a convenient model system to clarify the influence of voltage-dependent ion fluxes across PM on photosynthetic activity of chloroplasts. This study shows that permeation of MV to their target sites in chloroplasts is restricted by PM in resting cells, but MV easily passes through ionic channels opened during the PM depolarization. This gated permeation of MV gives rise to strong non-photochemical quenching, decrease in the effective quantum yield of linear electron flow, apparent O2 uptake, and, finally, the enhanced ROS production, as detected by the fluorescent probe dichlorofluorescein. Taken together, the results indicate that the AP generation in the presence of MV acts as trigger for instant redirection of photosynthetic linear electron flow from CO2-dependent route to the path of O2 reduction with the eventual formation of H2O2 as a dominant and most stable ROS form.


Subject(s)
Cell Membrane , Chara , Oxygen , Paraquat , Photosynthesis , Photosynthesis/drug effects , Electron Transport/drug effects , Paraquat/pharmacology , Cell Membrane/metabolism , Oxygen/metabolism , Chara/metabolism , Chara/drug effects , Oxidation-Reduction , Chloroplasts/metabolism
14.
Nat Commun ; 15(1): 7888, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39251596

ABSTRACT

Silica cell-wall formation in diatoms is a showcase for the ability of organisms to control inorganic mineralization. The process of silicification by these unicellular algae is tightly regulated within a membrane-bound organelle, the silica deposition vesicle (SDV). Two opposing scenarios were proposed to explain the tight regulation of this intracellular process: a template-mediated process that relies on preformed scaffolds, or a template-independent self-assembly process. The present work points to a third scenario, where the SDV membrane is a dynamic mold that shapes the forming silica. We use in-cell cryo-electron tomography to visualize the silicification process in situ, in its native-state, and with a nanometer-scale resolution. This reveals that the plasma membrane interacts with the SDV membrane via physical tethering at membrane contact sites, where the curvature of the tethered side of the SDV membrane mirrors the intricate silica topography. We propose that silica growth and morphogenesis result from the biophysical properties of the SDV and plasma membranes.


Subject(s)
Cell Membrane , Diatoms , Morphogenesis , Silicon Dioxide , Diatoms/metabolism , Diatoms/ultrastructure , Diatoms/growth & development , Silicon Dioxide/chemistry , Silicon Dioxide/metabolism , Cell Membrane/metabolism , Electron Microscope Tomography , Cell Wall/metabolism , Cell Wall/ultrastructure , Cryoelectron Microscopy
15.
ACS Appl Mater Interfaces ; 16(37): 49660-49672, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39240784

ABSTRACT

Atherosclerosis is a persistent inflammatory condition of the blood vessels associated with abnormalities in lipid metabolism. Development of biomimetic nanoplatforms provides an effective strategy. Herein, inspired by the peptide CLIKKPF spontaneously coupling to phosphatidylserine (PS) on the inner leaflet of cell membranes specifically, MM@NPs were constructed by macrophage membrane spontaneous encapsulation of cyclodextrin-based nanoparticles modified with the peptide CLIKKPF and loaded with the hydrophobic compound resveratrol. MM@NPs could be specifically phagocytized by the activated endothelium with the overexpressed VCAM-1 for enhancing target delivery into the pathological lesion. Additionally, for the ApoE-/- mice, MM@NPs provide comprehensive treatment efficiency in reducing oxidant stress, alleviating the inherent inflammation, and decreasing cholesterol deposition, subsequently resulting in the atherosclerotic plaque regression. Therefore, MM@NPs could be one possible candidate for improving lipid metabolism and inflammation in atherosclerosis.


Subject(s)
Atherosclerosis , Cyclodextrins , Inflammation , Lipid Metabolism , Macrophages , Nanoparticles , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Mice , Macrophages/metabolism , Macrophages/drug effects , Cyclodextrins/chemistry , Cyclodextrins/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism/drug effects , Nanoparticles/chemistry , RAW 264.7 Cells , Resveratrol/chemistry , Resveratrol/pharmacology , Nanomedicine , Cell Membrane/metabolism , Cell Membrane/drug effects , Humans
16.
J Neurosci Res ; 102(9): e25382, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39253877

ABSTRACT

Neurons establish functional connections responsible for how we perceive and react to the world around us. Communication from a neuron to its target cell occurs through a long projection called an axon. Axon distances can exceed 1 m in length in humans and require a dynamic microtubule cytoskeleton for growth during development and maintenance in adulthood. Stathmins are microtubule-associated proteins that function as relays between kinase signaling and microtubule polymerization. In this review, we describe the prolific role of Stathmins in microtubule homeostasis with an emphasis on emerging roles for Stathmin-2 (Stmn2) in axon integrity and neurodegeneration. Stmn2 levels are altered in Amyotrophic Lateral Sclerosis and loss of Stmn2 provokes motor and sensory neuropathies. There is growing potential for employing Stmn2 as a disease biomarker or even a therapeutic target. Meeting this potential requires a mechanistic understanding of emerging complexity in Stmn2 function. In particular, Stmn2 palmitoylation has a surprising contribution to axon maintenance through undefined mechanisms linking membrane association, tubulin interaction, and axon transport. Exploring these connections will reveal new insight on neuronal cell biology and novel opportunities for disease intervention.


Subject(s)
Axons , Microtubules , Stathmin , Stathmin/metabolism , Microtubules/metabolism , Humans , Axons/metabolism , Axons/physiology , Animals , Cell Membrane/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology
17.
Commun Biol ; 7(1): 1161, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289481

ABSTRACT

Oxidative stress in combination with acid stress has been shown to inactivate a wide spectrum of microorganisms, including multi-resistant bacteria. This occurs e.g. in phagolysosomes or during treatment by cold atmospheric pressure plasmas (CAP) and possibly depends on the cell membrane. We therefore explored the effects of CAP-generated reactive oxygen and nitrogen species (RONS) on bacterial growth inhibition and membranes in neutral and acidic suspensions. We observed that growth inhibition was most efficient when bacteria were treated by a mix of short and long-lived RONS in an acidic environment. Membrane packing was affected mainly upon contact with short-lived RONS, while also acidity strongly modulated packing. Under these conditions, Gram-negative bacteria displayed large potassium release while SYTOX Green influx remained marginal. Growth inhibition of Gram-negative bacteria correlated well with outer membrane (OM) permeabilization that occurred upon contact with short and/or long-lived RONS in synergy with acidity. In Gram-positive bacteria, CAP impaired membrane potential possibly through pore formation upon contact with short-lived RONS while formation of membrane protein hydroperoxides was probably involved in these effects. In summary, our study provides a wide perspective on understanding inactivation mechanisms of bacteria by RONS in combination with acidity.


Subject(s)
Cell Membrane , Escherichia coli , Oxidative Stress , Oxidative Stress/drug effects , Escherichia coli/drug effects , Escherichia coli/physiology , Escherichia coli/metabolism , Escherichia coli/growth & development , Cell Membrane/metabolism , Cell Membrane/drug effects , Staphylococcus/drug effects , Staphylococcus/physiology , Staphylococcus/metabolism , Staphylococcus/growth & development , Reactive Oxygen Species/metabolism , Acids/pharmacology , Acids/metabolism , Reactive Nitrogen Species/metabolism , Hydrogen-Ion Concentration , Plasma Gases/pharmacology
18.
Cell Chem Biol ; 31(9): 1627-1635, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303699

ABSTRACT

The dynamic process of membrane shaping and remodeling plays a vital role in cellular functions, with proteins and cellular membranes interacting intricately to adapt to various cellular needs and environmental cues. Ubiquitination-a posttranslational modification-was shown to be essential in regulating membrane structure and shape. It influences virtually all pathways relying on cellular membranes, such as endocytosis and autophagy by directing protein degradation, sorting, and oligomerization. Ubiquitin is mostly known as a protein modifier; however, it was reported that ubiquitin and ubiquitin-like proteins can associate directly with lipids, affecting membrane curvature and dynamics. In this review, we summarize some of the current knowledge on ubiquitin-mediated membrane remodeling in the context of endocytosis, autophagy, and ER-phagy.


Subject(s)
Cell Membrane , Ubiquitin , Ubiquitination , Ubiquitin/metabolism , Humans , Cell Membrane/metabolism , Autophagy , Endocytosis , Animals , Endoplasmic Reticulum/metabolism
19.
Nat Commun ; 15(1): 8214, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294175

ABSTRACT

CoQ10 (Coenzyme Q10) is an essential fat-soluble metabolite that plays a key role in cellular metabolism. A less-known function of CoQ10 is whether it may act as a plasma membrane-stabilizing agent and whether this property can affect cancer development and progression. Here, we show that CoQ10 and its biosynthetic enzyme UBIAD1 play a critical role in plasmamembrane mechanical properties that are of interest for breast cancer (BC) progression and treatment. CoQ10 and UBIAD1 increase membrane fluidity leading to increased cell stiffness in BC. Furthermore, CoQ10 and UBIAD1 states impair ECM (extracellular matrix)-mediated oncogenic signaling and reduce ferroptosis resistance in BC settings. Analyses on human patients and mouse models reveal that UBIAD1 loss is associated with BC development and progression and UBIAD1 expression in BC limits CTCs (circulating tumor cells) survival and lung metastasis formation. Overall, this study reveals that CoQ10 and UBIAD1 can be further investigated to develop therapeutic interventions to treat BC patients with poor prognosis.


Subject(s)
Breast Neoplasms , Extracellular Matrix , Ferroptosis , Signal Transduction , Ubiquinone , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism , Humans , Ferroptosis/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Animals , Female , Extracellular Matrix/metabolism , Mice , Cell Line, Tumor , Cell Membrane/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Gene Expression Regulation, Neoplastic
20.
Elife ; 122024 Sep 19.
Article in English | MEDLINE | ID: mdl-39297609

ABSTRACT

The Sonic hedgehog (Shh) signaling pathway controls embryonic development and tissue homeostasis after birth. This requires regulated solubilization of dual-lipidated, firmly plasma membrane-associated Shh precursors from producing cells. Although it is firmly established that the resistance-nodulation-division transporter Dispatched (Disp) drives this process, it is less clear how lipidated Shh solubilization from the plasma membrane is achieved. We have previously shown that Disp promotes proteolytic solubilization of Shh from its lipidated terminal peptide anchors. This process, termed shedding, converts tightly membrane-associated hydrophobic Shh precursors into delipidated soluble proteins. We show here that Disp-mediated Shh shedding is modulated by a serum factor that we identify as high-density lipoprotein (HDL). In addition to serving as a soluble sink for free membrane cholesterol, HDLs also accept the cholesterol-modified Shh peptide from Disp. The cholesteroylated Shh peptide is necessary and sufficient for Disp-mediated transfer because artificially cholesteroylated mCherry associates with HDL in a Disp-dependent manner, whereas an N-palmitoylated Shh variant lacking C-cholesterol does not. Disp-mediated Shh transfer to HDL is completed by proteolytic processing of the palmitoylated N-terminal membrane anchor. In contrast to dual-processed soluble Shh with moderate bioactivity, HDL-associated N-processed Shh is highly bioactive. We propose that the purpose of generating different soluble forms of Shh from the dual-lipidated precursor is to tune cellular responses in a tissue-type and time-specific manner.


Subject(s)
Hedgehog Proteins , Lipoproteins, HDL , Hedgehog Proteins/metabolism , Animals , Lipoproteins, HDL/metabolism , Mice , Humans , Cell Membrane/metabolism , Signal Transduction , Cholesterol/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL