Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (104)

Search Parameters:
Keywords = cellular-membrane-derived vesicles

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 7900 KB  
Article
Sublethal Doxorubicin Promotes Extracellular Vesicle Biogenesis in A375 Melanoma Cells: Implications for Vesicle-Loaded TGF-β-Mediated Cancer Progression and Cardiovascular Pathophysiology
by Laura Fernanda Fernández-Fonseca, Susana Novoa-Herrán, Adriana Umaña-Pérez and Luis Alberto Gómez-Grosso
Int. J. Mol. Sci. 2025, 26(17), 8524; https://doi.org/10.3390/ijms26178524 - 2 Sep 2025
Viewed by 620
Abstract
Doxorubicin (Dox) is not a first-line treatment for melanoma due to limited antitumor efficacy and dose-dependent cardiotoxicity. However, sublethal doses may trigger adaptive cellular responses that influence tumor progression and systemic toxicity. Small extracellular vesicles (EVs) are key mediators of intercellular communication and [...] Read more.
Doxorubicin (Dox) is not a first-line treatment for melanoma due to limited antitumor efficacy and dose-dependent cardiotoxicity. However, sublethal doses may trigger adaptive cellular responses that influence tumor progression and systemic toxicity. Small extracellular vesicles (EVs) are key mediators of intercellular communication and can carry bioactive molecules that modulate both the tumor microenvironment and distant tissues. This study investigates how sublethal Dox exposure alters EV biogenesis and cargo in A375 melanoma cells and explores the potential implications for cardiovascular function. We treated human A375 melanoma cells with 10 nM dox for 96 h. EVs were isolated using differential ultracentrifugation and size exclusion chromatography. Vesicle characterization included Immunocytochemistry for CD63, CD81, CD9, Rab7 and TSG101, scanning electron microscopy (SEM) Nanoparticle Tracking Analysis (NTA), and Western blotting for CD81 and CytC. We analyzed cytokine content using cytokine membrane arrays. Guinea pig cardiomyocytes were exposed to the isolated vesicles, and mitochondrial activity was evaluated using the MTT assay. Statistical analysis included t-tests, ANOVA, Cohen’s d, and R2 and η2. Dox exposure significantly increased EV production (13.6-fold; p = 0.000014) and shifted vesicle size distribution. CD81 expression was significantly upregulated (p = 0.0083), and SEM (microscopy) confirmed enhanced vesiculation. EVs from treated cells were enriched in TGF-β (p = 0.0134), VEGF, CXCL1, CXCL12, CCL5, IL-3, IL-4, IL-10, Galectin-3, and KITLG. Cardiomyocytes exposed to these vesicles showed a 2.3-fold reduction in mitochondrial activity (p = 0.0021), an effect absent when vesicles were removed. Bioinformatic analysis linked EV cargo to pathways involved in cardiac hypertrophy, inflammation, and fibrosis. As conclusion, sublethal Doxorubicin reprograms melanoma-derived EVs by enhancing their production and enriching their cargo with profibrotic and immunomodulatory mediators. These vesicles may contribute to tumor progression and cardiovascular physiopathology, suggesting that targeting EVs could improve therapeutic outcomes in cancer and cardiovascular disease. Full article
Show Figures

Figure 1

15 pages, 2702 KB  
Article
Evaluation of Hypovirus Infection on the Vesicular Protein Expression Pattern of Cryphonectria parasitica by TMT-Based Proteomics Analysis
by Zishan Zeng, Nanxin Lin, Tao Lu, Jian Xu, Zheng Zhang, Fang Wang and Jinzi Wang
Biology 2025, 14(9), 1123; https://doi.org/10.3390/biology14091123 - 25 Aug 2025
Viewed by 529
Abstract
Hypovirus infection is known to reduce the pathogenicity of Cryphonectria parasitica, the causative agent of chestnut blight. Isoforms derived from a viral protein p48 have been discovered in host mitochondria and vesicles, which may contribute to virulence attenuation, as reported in earlier [...] Read more.
Hypovirus infection is known to reduce the pathogenicity of Cryphonectria parasitica, the causative agent of chestnut blight. Isoforms derived from a viral protein p48 have been discovered in host mitochondria and vesicles, which may contribute to virulence attenuation, as reported in earlier work using two-dimensional electrophoresis (2-DE). In this study, a total of 1739 fungal proteins were identified in fungal vesicles through Tandem Mass Tag (TMT)-based quantitative proteomics. The infection of CHV1-EP713 was associated with 75 up-regulated and 201 down-regulated proteins, predominantly involved in vesicular transport process and related cellular functions, including protein folding, membrane fusion, retrograde transport, autophagy, and ER stress responses. The down-regulation of calnexin, COPI, ArfGAP, importin-β, and Atg8 is consistent with impairments in protein folding, retrograde transport, and autophagy. Meanwhile, the up-regulation of clathrin, dynamin, Vps10p, HSP70, and t-SNAREs indicated enhanced trafficking to vacuoles and increased stress response activity. Overall, our findings indicate that hypoviral infection is associated with extensive alterations in the vesicular transport system of C. parasitica, likely mediated through changes in the abundance of multiple key protein regulators. These alterations may underlie attenuation of virulence by impacting crucial cellular processes. Full article
(This article belongs to the Section Infection Biology)
Show Figures

Graphical abstract

43 pages, 8518 KB  
Review
Cutting-Edge Sensor Technologies for Exosome Detection: Reviewing Role of Antibodies and Aptamers
by Sumedha Nitin Prabhu and Guozhen Liu
Biosensors 2025, 15(8), 511; https://doi.org/10.3390/bios15080511 - 6 Aug 2025
Viewed by 1626
Abstract
Exosomes are membranous vesicles that play a crucial role as intercellular messengers. Cells secrete exosomes, which can be found in a variety of bodily fluids such as amniotic fluid, semen, breast milk, tears, saliva, urine, blood, bile, ascites, and cerebrospinal fluid. Exosomes have [...] Read more.
Exosomes are membranous vesicles that play a crucial role as intercellular messengers. Cells secrete exosomes, which can be found in a variety of bodily fluids such as amniotic fluid, semen, breast milk, tears, saliva, urine, blood, bile, ascites, and cerebrospinal fluid. Exosomes have a distinct bilipid protein structure and can be as small as 30–150 nm in diameter. They may transport and exchange multiple cellular messenger cargoes across cells and are used as a non-invasive biomarker for various illnesses. Due to their unique features, exosomes are recognized as the most effective biomarkers for cancer and other disease detection. We give a review of the most current applications of exosomes derived from various sources in the prognosis and diagnosis of multiple diseases. This review also briefly examines the significance of exosomes and their applications in biomedical research, including the use of aptamers and antibody–antigen functionalized biosensors. Full article
(This article belongs to the Special Issue Material-Based Biosensors and Biosensing Strategies)
Show Figures

Figure 1

15 pages, 1027 KB  
Article
Enhanced Outer Membrane Vesicle Production in Escherichia coli: From Metabolic Network Model to Designed Strain Lipidomic Profile
by Héctor Alejandro Ruiz-Moreno, Juan D. Valderrama-Rincon, Mónica P. Cala, Miguel Fernández-Niño, Mateo Valderruten Cajiao, María Francisca Villegas-Torres and Andrés Fernando González Barrios
Int. J. Mol. Sci. 2025, 26(14), 6714; https://doi.org/10.3390/ijms26146714 - 13 Jul 2025
Cited by 1 | Viewed by 1016
Abstract
Bacterial structures formed from the outer membrane and the periplasm components carry biomolecules to expel cellular material and interact with other cells. These outer membrane vesicles (OMVs) can encapsulate bioactive content, which confers OMVs with high potential as alternative drug delivery vehicles or [...] Read more.
Bacterial structures formed from the outer membrane and the periplasm components carry biomolecules to expel cellular material and interact with other cells. These outer membrane vesicles (OMVs) can encapsulate bioactive content, which confers OMVs with high potential as alternative drug delivery vehicles or as a platform for novel vaccine development. Single-gene mutants derived from Escherichia coli JC8031 were engineered to further enhance OMV production based on metabolic network modelling and in silico gene knockout design (ΔpoxB, ΔsgbE, ΔgmhA, and ΔallD). Mutants were experimentally obtained by genome editing using CRISPR-Cas9 and tested for OMVs recovery observing an enhanced OMV production in all of them. Lipidomic analysis through LC-ESI-QTOF-MS was performed for OMVs obtained from each engineered strain and compared to the wild-type E. coli JC8031 strain. The lipid profile of OMVs from the wild-type E. coli JC8031 did not change significantly confirmed by multivariate statistical analysis when compared to the mutant strains. The obtained results suggest that the vesicle production can be further improved while the obtained vesicles are not altered in their composition, allowing further study for stability and integrity for use in therapeutic settings. Full article
(This article belongs to the Special Issue From Molecular to Systems Biology through Data Integration)
Show Figures

Figure 1

26 pages, 6136 KB  
Review
Exosomes as Future Therapeutic Tools and Targets for Corneal Diseases
by Joshua Gamez, Daxian Zha, Shaghaiegh M. Ebrahimi, Seok White, Alexander V. Ljubimov and Mehrnoosh Saghizadeh
Cells 2025, 14(13), 959; https://doi.org/10.3390/cells14130959 - 23 Jun 2025
Viewed by 1806
Abstract
The therapeutic potential of exosomes (Exos), a subpopulation of extracellular vesicles (EVs) secreted by various cell types, has been broadly emphasized. Exos are endosome-derived membrane-bound vesicles 50–150 nm in size. Exos can be general or cell type-specific. Their contents enable them to function [...] Read more.
The therapeutic potential of exosomes (Exos), a subpopulation of extracellular vesicles (EVs) secreted by various cell types, has been broadly emphasized. Exos are endosome-derived membrane-bound vesicles 50–150 nm in size. Exos can be general or cell type-specific. Their contents enable them to function as multi-signaling and vectorized vehicles. Exos are important for maintaining cellular homeostasis. They are released into extracellular spaces, leading to uptake by neighboring or distant cells and delivering their contents to modulate cell signaling. Exos influence tissue responses to injury, infection, and disease by fusion with the target cells and transferring their cargo, including cytokines, growth and angiogenic factors, signaling molecules, lipids, DNA, mRNAs, and non-coding RNAs. They are implicated in various physiological and pathological conditions, including ocular surface events, such as corneal scarring, wound healing, and inflammation. Their biocompatibility, stability, low immunogenicity, and easy detectability in bodily fluids (blood, tears, saliva, and urine) make them promising tools for diagnosing and treating ocular diseases. The potential to engineer specific Exo cargos makes them outstanding therapeutic delivery vehicles. The objective of this review is to provide novel insights into the functions of Exo cargos and their applications as biomarkers and therapeutics, or targets in the cornea. Full article
Show Figures

Figure 1

17 pages, 5473 KB  
Article
Sivelestat-Loaded Neutrophil-Membrane-Coated Antioxidative Nanoparticles for Targeted Endothelial Protection in Sepsis
by Juexian Wei, Aijia Zhong, Yuting Zhang, Ehua Deng, Hengzong Mo, Hongyu Zhao, Jiayu Huang, Huaidong Peng, Kaiyin Zhang, Xiaohui Chen, Haifeng Mao, Yixin Chen and Yongcheng Zhu
Pharmaceutics 2025, 17(6), 766; https://doi.org/10.3390/pharmaceutics17060766 - 10 Jun 2025
Viewed by 1107
Abstract
Background/Objectives: This study aims to develop and evaluate neutrophil-membrane-coated nanoparticles (Siv@NMs) encapsulating sivelestat for the treatment of sepsis-induced endothelial injury. Leveraging the intrinsic chemotactic properties of neutrophil membranes, Siv@NMs are engineered to achieve site-specific delivery of sivelestat to damaged endothelia, thereby overcoming [...] Read more.
Background/Objectives: This study aims to develop and evaluate neutrophil-membrane-coated nanoparticles (Siv@NMs) encapsulating sivelestat for the treatment of sepsis-induced endothelial injury. Leveraging the intrinsic chemotactic properties of neutrophil membranes, Siv@NMs are engineered to achieve site-specific delivery of sivelestat to damaged endothelia, thereby overcoming the limitations of conventional therapies in mitigating endothelial dysfunction and multiorgan failure associated with sepsis. Methods: Siv@NMs were synthesized through a combination of ultrasonication and extrusion techniques to encapsulate sivelestat within neutrophil-membrane-derived vesicles. Comprehensive physicochemical characterization included analysis of particle size distribution, zeta potential, and encapsulation efficiency. Stability profiles and controlled release kinetics were systematically evaluated under simulated conditions. In vitro investigations encompassed (1) endothelial cell biocompatibility assessment via cytotoxicity assays, (2) investigation of the targeting efficiency in suppressing endothelial neutrophil extracellular trap generation during inflammation, and (3) ROS-scavenging capacity quantification using flow cytometry with DCFH-DA fluorescent probes. In vivo therapeutic efficacy was validated using a cecal ligation and puncture (CLP) sepsis mouse model, with multiparametric monitoring of endothelial function, inflammatory markers, ROS levels, and survival outcomes. Results: The optimized Siv@NMs exhibited an average particle size of approximately 150 nm, and a zeta potential of −10 mV was achieved. Cellular studies revealed that (1) Siv@NMs selectively bound to inflammatory endothelial cells with minimal cytotoxicity, and (2) Siv@NMs significantly reduced ROS accumulation in endothelial cells subjected to septic stimuli. In vitro experiments demonstrated that Siv@NMs treatment markedly attenuated endothelial injury biomarkers’ expression (ICAM-1 and iNOS), suppressed formation of neutrophil extracellular traps, and improved survival rates compared to treatment with free sivelestat. Conclusions: The neutrophil-membrane-coated nanoparticles loaded with sivelestat present a breakthrough strategy for precision therapy of sepsis-associated endothelial injury. This bioengineered system synergistically combines targeted drug delivery with multimodal therapeutic effects, including ROS mitigation, anti-inflammatory action, and endothelial protection. These findings substantiate the clinical translation potential of Siv@NMs as a next-generation nanotherapeutic for sepsis management. Full article
(This article belongs to the Special Issue ROS-Mediated Nano Drug Delivery for Antitumor Therapy)
Show Figures

Figure 1

18 pages, 4464 KB  
Article
Protective Effect of Mesenchymal Stem Cell-Derived Extracellular Vesicles on Inner Ear Sensorineural Cells Affected by Cisplatin
by Maria Perde-Schrepler, Ioana Brie, Mihai Cenariu, Sergiu Chira, Lajos Raduly, Liviuta Budisan, Ioana Berindan-Neagoe, Rares Stiufiuc, Maximilian Dindelegan, Cristina Blebea, Emoke Pall and Alma Aurelia Maniu
Medicina 2025, 61(6), 1042; https://doi.org/10.3390/medicina61061042 - 5 Jun 2025
Viewed by 821
Abstract
Background and Objectives: Extracellular vesicles (EVs) derived from mesenchymal stem cells have gained much attention as potential therapeutic agents in many diseases, including hearing disorders such as sensorineural hearing loss (SNHL). EVs inherit similar therapeutic effects, including the stimulation of tissue regeneration [...] Read more.
Background and Objectives: Extracellular vesicles (EVs) derived from mesenchymal stem cells have gained much attention as potential therapeutic agents in many diseases, including hearing disorders such as sensorineural hearing loss (SNHL). EVs inherit similar therapeutic effects, including the stimulation of tissue regeneration from the parental cells. The aim of our study was to isolate EVs produced by MSCs and use them to treat inner ear cells in culture to evaluate their protective potential against the damaging effect of an ototoxic drug. Materials and Methods: We isolated MSC-derived EVs by precipitation and characterized them by number, size, and morphology using nanoparticle tracking analysis and TEM, evaluated the protein concentration by BCA assay and the presence of EV markers CD9, CD63, and CD81 by the Dot Blot immunoblotting method. HEI-OC1 inner ear cell line was treated with EVs either alone or followed by Cisplatin. We assessed the uptake of EVs in HEI-OC1 cells by fluorescence microscopy after PKH26 labeling, ROS production by the DCFDA (dichlorfluorescein diacetate) assay, cellular viability by Alamar Blue assay, and apoptosis with the Annexin V/Propidium Iodide method. Results: The isolated EVs had mean dimensions of 184.4 nms and the concentration of the EV suspension was 180 × 106 particles/mL. TEM analysis showed intact vesicular structures with lipid-bilayer membranes having similar sizes with those measured by NTA. The PKH26-labeled EVs were observed in the HEI-OC1 cells after 24 h incubation, the amount increasing with the concentration. EVs reduced ROS production and increased the number of viable cells both alone and as pretreatment before Cisplatin, dose-dependently. Cells in early apoptosis were inhibited by EVs, while those in late apoptosis were enhanced, both with and without Cisplatin. Conclusions: EVs secreted by MSC protected HEI-OC1 cells against Cisplatin toxicity, reduced ROS production, and stimulated cell viability and the elimination of damaged cells by apoptosis, protecting the HEI-OC1 cells against Cisplatin-induced damage. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

16 pages, 512 KB  
Review
The Role of Helicobacter pylori Heat Shock Proteins in Gastric Diseases’ Pathogenesis
by Olga Maria Manna, Celeste Caruso Bavisotto, Melania Ionelia Gratie, Provvidenza Damiani, Giovanni Tomasello and Francesco Cappello
Int. J. Mol. Sci. 2025, 26(11), 5065; https://doi.org/10.3390/ijms26115065 - 24 May 2025
Cited by 2 | Viewed by 3164
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative bacterium that colonizes the human stomach and is associated with several gastric diseases, including gastritis, peptic ulcer disease, and gastric cancer. The bacterium’s ability to thrive in the harsh gastric environment is due, to [...] Read more.
Helicobacter pylori (H. pylori) is a Gram-negative bacterium that colonizes the human stomach and is associated with several gastric diseases, including gastritis, peptic ulcer disease, and gastric cancer. The bacterium’s ability to thrive in the harsh gastric environment is due, to some extent, to its stress response mechanisms, with its heat shock proteins (HSPs) playing a putative, yet not fully understood, role in these adaptive processes. HSPs are a family of molecules, highly conserved throughout phylogenesis, that assist in protein folding, prevent aggregation, and ensure cellular homeostasis under stressful conditions. In H. pylori, HSPs contribute to survival in the stomach’s acidic environment and oxidative stress. Furthermore, they aid in the bacterium’s ability to adhere to gastric epithelial cells, modulate the host immune response, and form biofilms, all contributing to chronic infection and pathogenicity. The role of microbial HSPs in antibiotic resistance has also emerged as a critical area of research, as these proteins help stabilize efflux pumps, protect essential proteins targeted by antibiotics, and promote biofilm formation, thereby reducing the efficacy of antimicrobial treatments. Among bacterial HSPs, GroEL and DnaK are probably the major proteins that control most of the H. pylori’s functioning. Indeed, both proteins possess remarkable acid resistance, high substrate affinity, and dual roles in protein homeostasis and host interaction. These features make them critical for H. pylori’s adaptation, persistence, and pathogenicity in the gastric niche. In addition, recent findings have also highlighted the involvement of HSPs in the crosstalk between H. pylori and gastric epithelial cells mediated by the release of bacterial outer membrane vesicles and host-derived exosomes, both of these extracellular vesicles being part of the muco-microbiotic layer of the stomach and influencing cellular signalling and immune modulation. Considering their critical role in the survival and persistence of bacteria, microbial HSPs also represent potential therapeutic targets. Strategies aimed at inhibiting microbial HSP function, combined with conventional antibiotics or developing vaccines targeting microbial HSPs, could provide new avenues for the treatment of H. pylori infections and combat antibiotic resistance. This review explores the multifaceted roles of microbial HSPs in the pathogenesis of H. pylori, highlighting their contributions to bacterial adhesion, immune evasion, stress response, and antibiotic resistance. Full article
(This article belongs to the Special Issue Pathogenicity and Antibiotic Resistance of Helicobacter pylori)
Show Figures

Graphical abstract

19 pages, 2623 KB  
Article
Phytyl Phenolipids: Structurally Modified Antioxidants with Superior Lipid Membrane Interaction
by Vânia Costa, Marlene Costa, Rute Rebelo, Francisca Arques, Mariana Ferreira, Paula Gameiro, Tomás Barros, Dulce Geraldo, Luís S. Monteiro and Fátima Paiva-Martins
Molecules 2025, 30(10), 2193; https://doi.org/10.3390/molecules30102193 - 17 May 2025
Cited by 1 | Viewed by 684
Abstract
A set of procedures was developed for the simple synthesis of phytyl phenolipids, which resulted in high yields (70–95%) of phytyl esters of caffeic, protocatechuic, homoprotocatechuic, and dihydrocaffeic acids. Initial characterization revealed that these new compounds exhibited similar radical scavenging activity and liposolubility [...] Read more.
A set of procedures was developed for the simple synthesis of phytyl phenolipids, which resulted in high yields (70–95%) of phytyl esters of caffeic, protocatechuic, homoprotocatechuic, and dihydrocaffeic acids. Initial characterization revealed that these new compounds exhibited similar radical scavenging activity and liposolubility to α-tocopherol, a key antioxidant present in membranes. Cyclic voltammetry analysis indicated that the phytyl derivatives had lower anodic peak potentials compared to the original phenolic acids, with electron transfer following an adsorption-controlled mechanism. In phosphatidylcholine large unilamellar vesicles (LUVs), phytyl esters demonstrated remarkable efficiency in preventing liposome autoxidation when compared to α-tocopherol. Despite their strong radical scavenging capacity and membrane penetration ability, the antioxidant effectiveness of the phytyl esters in liposomes was influenced by the structure of their polyphenolic moiety. These new compounds are considered promising candidates for future pharmacological applications against oxidative stress in lipoproteins and cells, warranting further evaluation of their antioxidant and anti-inflammatory effects in cellular models and in vivo. Full article
(This article belongs to the Section Macromolecular Chemistry)
Show Figures

Figure 1

24 pages, 1026 KB  
Review
Advances in Plant-Derived Extracellular Vesicle Extraction Methods and Pharmacological Effects
by Nuerbiye Nueraihemaiti, Dilihuma Dilimulati, Alhar Baishan, Sendaer Hailati, Nulibiya Maihemuti, Alifeiye Aikebaier, Yipaerguli Paerhati and Wenting Zhou
Biology 2025, 14(4), 377; https://doi.org/10.3390/biology14040377 - 6 Apr 2025
Cited by 2 | Viewed by 2750
Abstract
Extracellular vesicles (EVs) are those with a double-membrane structure that contains proteins, lipids, nucleic acids, and other biologically active substances that play an important role in cell–cell and cell–environment communication. They have also become an important mechanism for exchanging biologically active substances for [...] Read more.
Extracellular vesicles (EVs) are those with a double-membrane structure that contains proteins, lipids, nucleic acids, and other biologically active substances that play an important role in cell–cell and cell–environment communication. They have also become an important mechanism for exchanging biologically active substances for cellular molecules. As many studies on EVs have been conducted, plant-derived extracellular vesicles (PDEVs) have also started attracting attention. The biological activity and stability of PDEVs are closely related to the extraction and separation methods, and choosing a separation method that meets the requirements of PDEVs is important. The extraction methods of PDEVs include ultracentrifugation, ultrafiltration, size-exclusion chromatography, etc. In recent years, it has been found through research that PDEVs possess biological properties, such as anti-inflammatory, anti-cancer, and anti-infective properties, and that they show unique advantages as therapeutic agents and drug carriers. Therefore, we have collected the scientific literature related to EVs derived from more than a dozen fruits and vegetables, and summarized and analyzed their extraction, separation, and roles in disease treatment, aiming to provide reference and inspiration for the in-depth study of the efficacy of new drugs. Full article
(This article belongs to the Section Biotechnology)
Show Figures

Graphical abstract

15 pages, 857 KB  
Review
Innate Immunity in Cystic Fibrosis: Varied Effects of CFTR Modulator Therapy on Cell-to-Cell Communication
by Jennifer Hynes, Clifford C. Taggart, Rabindra Tirouvanziam and Judith A. Coppinger
Int. J. Mol. Sci. 2025, 26(6), 2636; https://doi.org/10.3390/ijms26062636 - 14 Mar 2025
Cited by 1 | Viewed by 1623
Abstract
Cystic Fibrosis (CF) is a life-shortening, multi-organ disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Prominent clinical features of CF take place in the lung, hallmarked by cycles of bacterial infection and a dysfunctional inflammatory airway response, leading to [...] Read more.
Cystic Fibrosis (CF) is a life-shortening, multi-organ disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Prominent clinical features of CF take place in the lung, hallmarked by cycles of bacterial infection and a dysfunctional inflammatory airway response, leading to eventual respiratory failure. Bidirectional crosstalk between epithelial cells, leukocytes (e.g., neutrophils, macrophages) and bacteria via release of intra-cellular mediators is key to driving inflammation in CF airways. In recent years, a highly effective combination of therapeutics targeting the CFTR defect have revolutionized treatment in CF. Despite these advancements and due to the complexity of the immune response in the CF airway, the full impact of highly effective modulator therapy (HEMT) on airway inflammation is not fully determined. This review provides the evidence to date on crosstalk mechanisms between host epithelium, leukocytes and bacteria and examines the effect of HEMT on both soluble and membrane-derived immune mediators in clinical samples. The varied effects of HEMT on expression of key proteases, cytokines and extracellular vesicles (EVs) in relation to clinical parameters is assessed. Advances in treatment with HEMT have shown potential in dampening the chronic inflammatory response in CF airways. However, to fully quell inflammation and maximize lung tissue resilience, further interventions may be necessary. Exploring the effects of HEMT on key immune mediators paves the way for identifying new anti-inflammatory approaches targeting host immune cell interactions, such as EV-directed lung therapies. Full article
(This article belongs to the Special Issue New Research Insights in Cystic Fibrosis and CFTR-Related Diseases)
Show Figures

Figure 1

32 pages, 10992 KB  
Article
Small Extracellular Vesicles from Breast Cancer Cells Induce Cardiotoxicity
by Jhon Jairo Osorio-Méndez, Luis Alberto Gómez-Grosso, Gladis Montoya-Ortiz, Susana Novoa-Herrán and Yohana Domínguez-Romero
Int. J. Mol. Sci. 2025, 26(3), 945; https://doi.org/10.3390/ijms26030945 - 23 Jan 2025
Cited by 2 | Viewed by 2573
Abstract
Cardiovascular diseases and cancer are leading global causes of morbidity and mortality, necessitating advances in diagnosis and treatment. Doxorubicin (Doxo), a potent chemotherapy drug, causes long-term heart damage due to cardiotoxicity. Small extracellular vesicles (sEVs) carry bioactive molecules—such as proteins, lipids, and nucleic [...] Read more.
Cardiovascular diseases and cancer are leading global causes of morbidity and mortality, necessitating advances in diagnosis and treatment. Doxorubicin (Doxo), a potent chemotherapy drug, causes long-term heart damage due to cardiotoxicity. Small extracellular vesicles (sEVs) carry bioactive molecules—such as proteins, lipids, and nucleic acids—that can modulate gene expression and signaling pathways in recipient cells, including cardiomyocytes. Through the delivery of cytokines, microRNAs, and growth factors, sEVs can influence cell survival, which plays a critical role in the development of cardiotoxicity. This study investigates the role of sEVs derived from breast cancer cells treated or not with Doxo and their potential to induce cardiomyocyte damage, thereby contributing to cardiotoxicity. We isolated sEVs from MCF-7 cells treated or not to Doxo using ultracentrifugation and characterized them through Nanoparticle Tracking Analysis (NTA), Scanning Electron Microscopy (SEM), and Western Blotting (WB) for the markers CD63, CD81, and TSG101. We analyzed cytokine profiles using a Multiplex Assay and Cytokine Membrane Array. We exposed Guinea pig cardiomyocytes to different concentrations of sEVs. We assessed their viability (MTT assay), shortening, reactive oxygen species (ROS–DHE dye) production, mitochondrial membrane potential (JC-1 dye), and calcium dynamics (FLUO-4 dye). We performed statistical analyses, including t-tests, ANOVA, Cohen’s d, and η2 to validate the robustness of the results. Treatment of MCF-7 cells with 0.01 μM Doxorubicin resulted in increased sEVs production, particularly after 48 h of exposure (~1.79 × 108 ± 2.77 × 107 vs. ~5.1 × 107 ± 1.28 × 107 particles/mL, n = 3, p = 0.0019). These sEVs exhibited protein profiles in the 130–25 kDa range and 93–123 nm sizes. They carried cytokines including TNF-α, IL-1β, IL-4, IFN-γ, and IL-10. Exposure of cardiomyocytes to sEVs (0.025 μg/mL to 2.5 μg/mL) from both Doxo-treated and untreated cells significantly reduced cardiomyocyte viability, shortened cell length by up to 20%, increased ROS production, and disrupted calcium homeostasis and mitochondrial membrane potential, indicating severe cellular stress and cardiotoxicity. These findings suggest that Doxo enhances sEVs production from breast cancer cells, which plays a key role in cardiotoxicity through their cytokine cargo. The study highlights the potential of these sEVs as biomarkers for early cardiotoxicity detection and as therapeutic targets to mitigate cardiovascular risks in chemotherapy patients. Future research should focus on understanding the mechanisms by which Doxorubicin-induced sEVs contribute to cardiotoxicity and exploring their diagnostic and therapeutic potential to improve patient safety and outcomes in cancer therapy. Full article
(This article belongs to the Special Issue Exosomes and Non-Coding RNA Research in Health and Disease)
Show Figures

Figure 1

31 pages, 1806 KB  
Review
Emerging Strategies for Revascularization: Use of Cell-Derived Extracellular Vesicles and Artificial Nanovesicles in Critical Limb Ischemia
by Vijay Murali Ravi Mythili, Ramya Lakshmi Rajendran, Raksa Arun, Vasanth Kanth Thasma Loganathbabu, Danyal Reyaz, ArulJothi Kandasamy Nagarajan, Byeong-Cheol Ahn and Prakash Gangadaran
Bioengineering 2025, 12(1), 92; https://doi.org/10.3390/bioengineering12010092 - 20 Jan 2025
Cited by 3 | Viewed by 2372
Abstract
Critical limb ischemia (CLI) poses a substantial and intricate challenge in vascular medicine, necessitating the development of innovative therapeutic strategies to address its multifaceted pathophysiology. Conventional revascularization approaches often fail to adequately address the complexity of CLI, necessitating the identification of alternative methodologies. [...] Read more.
Critical limb ischemia (CLI) poses a substantial and intricate challenge in vascular medicine, necessitating the development of innovative therapeutic strategies to address its multifaceted pathophysiology. Conventional revascularization approaches often fail to adequately address the complexity of CLI, necessitating the identification of alternative methodologies. This review explores uncharted territory beyond traditional therapies, focusing on the potential of two distinct yet interrelated entities: cell-derived extracellular vesicles (EVs) and artificial nanovesicles. Cell-derived EVs are small membranous structures naturally released by cells, and artificial nanovesicles are artificially engineered nanosized vesicles. Both these vesicles represent promising avenues for therapeutic intervention. They act as carriers of bioactive cargo, including proteins, nucleic acids, and lipids, that can modulate intricate cellular responses associated with ischemic tissue repair and angiogenesis. This review also assesses the evolving landscape of CLI revascularization through the unique perspective of cell-derived EVs and artificial nanovesicles. The review spans the spectrum from early preclinical investigations to the latest translational advancements, providing a comprehensive overview of the current state of research in this emerging field. These groundbreaking vesicle therapies hold immense potential for revolutionizing CLI treatment paradigms. Full article
(This article belongs to the Special Issue Innovations in Regenerative Therapy: Cell and Cell-Free Approaches)
Show Figures

Graphical abstract

18 pages, 5723 KB  
Article
Development and Biological Characterization of Cancer Biomimetic Membrane Nanovesicles for Enhancing Therapy Efficacy in Human Glioblastoma Cells
by Martina Massarotti, Paola Corna, Aromita Mallik, Gloria Milanesi, Claudio Casali, Lorenzo Magrassi and Sergio Comincini
Nanomaterials 2024, 14(22), 1779; https://doi.org/10.3390/nano14221779 - 5 Nov 2024
Cited by 1 | Viewed by 1396
Abstract
As nanocarriers of a new generation, biomimetic nanovesicles are an emerging class of therapeutic tools whose surface is integrated or fabricated with biomaterials capable of mimicking the biological features and functions of native cells. Thanks to this, biomimetic nanovesicles, in particular, those made [...] Read more.
As nanocarriers of a new generation, biomimetic nanovesicles are an emerging class of therapeutic tools whose surface is integrated or fabricated with biomaterials capable of mimicking the biological features and functions of native cells. Thanks to this, biomimetic nanovesicles, in particular, those made by plasma membrane moieties, possess greatly improved biocompatibility, high target specificity, a long retention time, and minimal undesired immune responses. For these reasons, a multitude of progenitor cells including cancer ones were employed as templates to generate biomimetic or membrane-camouflaged nanovesicles hosting different therapeutic compounds. In this contribution, different membrane-derived biomimetic vesicles (M-NVs) were generated by osmotic lysis or plasma membrane isolation approaches from normal and cancer cell lines and assayed against in vitro models of human glioblastoma. M-NVs were compared in their cellular internalization degrees of DNA and proteins, morphologically and molecularly characterized, expressing an extracellular membrane-associated marker. Then, Rose Bengal (RB), a photoactivable drug characterized by a relatively low cellular uptake, was incorporated into nascent glioblastoma-derived M-NVs and finally administered to homotypic receiving cells, showing an increased degree of internalization as well as induced cytotoxic effects, even in the absence of photodynamic direct stimulation. Similar results were also obtained assaying lyophilized M-NVs loaded with RB. In conclusion, M-NVs generated by cell membranes effectively deliver several cargoes, including therapeutic molecules, maintain functionality after lyophilization, and show significant internalization effects, making them a promising strategy for therapeutic applications against human glioblastoma cells. Full article
(This article belongs to the Special Issue The Study of the Effects of Nanoparticles on Human Cells)
Show Figures

Figure 1

15 pages, 2684 KB  
Article
Effect of Unfractionated Heparin Dose on Complement Activation and Selected Extracellular Vesicle Populations during Extracorporeal Membrane Oxygenation
by Johannes Zipperle, Laurenz Vock, Gerhard Fritsch, Johannes Grillari, Marcin F. Osuchowski, Wolfgang Holnthoner, Herbert Schöchl, Rebecca Halbgebauer, Markus Huber-Lang, Nikolaus Hofmann, Vincenz Scharner, Mauro Panigada, Johannes Gratz and Giacomo Iapichino
Int. J. Mol. Sci. 2024, 25(20), 11166; https://doi.org/10.3390/ijms252011166 - 17 Oct 2024
Cited by 1 | Viewed by 2639
Abstract
Extracorporeal membrane oxygenation (ECMO) provides critical support for patients with severe cardiopulmonary dysfunction. Unfractionated heparin (UFH) is used for anticoagulation to maintain circuit patency and avoid thrombotic complications, but it increases the risk of bleeding. Extracellular vesicles (EVs), nano-sized subcellular spheres with potential [...] Read more.
Extracorporeal membrane oxygenation (ECMO) provides critical support for patients with severe cardiopulmonary dysfunction. Unfractionated heparin (UFH) is used for anticoagulation to maintain circuit patency and avoid thrombotic complications, but it increases the risk of bleeding. Extracellular vesicles (EVs), nano-sized subcellular spheres with potential pro-coagulant properties, are released during cellular stress and may serve as potential targets for monitoring anticoagulation, particularly in thromboinflammation. We investigated the impact of UFH dose during ECMO therapy at the coagulation–inflammation interface level, focusing on complement activation and changes in circulating large EV (lEV) subsets. In a post hoc analysis of a multicenter randomized controlled trial comparing two anticoagulation management algorithms, we examined lEV levels and complement activation in 23 veno-venous-ECMO patients stratified by UFH dose. Blood samples were collected at different time points and grouped into three phases of ECMO therapy: initiation (day 1), mid (days 3–4), and late (days 6–7). Immunoassays detected complement activation, and flow cytometry analyzed lEV populations with an emphasis on mitochondria-carrying subsets. Patients receiving <15 IU/kg/h UFH exhibited higher levels of the complement activation product C5a and soluble terminal complement complex (sC5b-9). Lower UFH doses were linked to increased endothelial-derived lEVs, while higher doses were associated with elevated RBC-derived and mitochondria-positive lEVs. Our findings suggest the potential theranostic relevance of EV detection at the coagulation–inflammation interface. Further research is needed to standardize EV detection methods and validate these findings in larger ECMO patient cohorts. Full article
(This article belongs to the Special Issue Characterization of Extracellular Vesicles in Disease)
Show Figures

Figure 1

Back to TopTop