Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (88)

Search Parameters:
Keywords = modulated cargo release

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 6136 KiB  
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 546
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

21 pages, 1640 KiB  
Article
Analysis of Proteins and Piwi-Interacting RNA Cargo of Extracellular Vesicles (EVs) Isolated from Human Nose Organoids and Nasopharyngeal Secretions of Children with RSV Infections
by Tiziana Corsello, Nicholas Dillman, Yingxin Zhao, Teodora Ivanciuc, Tianshuang Liu, Antonella Casola and Roberto P. Garofalo
Viruses 2025, 17(6), 764; https://doi.org/10.3390/v17060764 - 28 May 2025
Viewed by 669
Abstract
Respiratory syncytial virus (RSV) is the leading cause of respiratory infections in children. Extracellular vesicles (EVs), released by airway epithelial cells, contain proteins and different families of non-coding RNAs (EV cargo) that can modulate the responses of target cells to viral infection. Nasal [...] Read more.
Respiratory syncytial virus (RSV) is the leading cause of respiratory infections in children. Extracellular vesicles (EVs), released by airway epithelial cells, contain proteins and different families of non-coding RNAs (EV cargo) that can modulate the responses of target cells to viral infection. Nasal mucosa is a primary site of viral entry and the source of EVs present in the upper airway secretions. In this study we characterized proteins, including inflammatory mediators and cytokines, and the piwi-interacting RNA (piRNAs) cargo of EVs isolated from pediatric human nose organoids (HNO) and nasopharyngeal secretions (NPS) positive for RSV. Using Proximity Extension Assay (PEA) and Luminex multi-target arrays, we found significant enrichment in several chemokines and other mediators/biomarkers, including CCL2, CCL20, CXCL5, CX3CL1, CXCL6, MMP-1, MMP-10, uPA, Flt3L, ARNT and CD40 in EVs secreted by RSV-infected HNO compared to control mock HNO. Analysis of NPS samples from RSV infected children revealed that CCL3, CCL20, CXCL8, uPA, VEGFA, were concentrated in the NPS-EV fraction. LC-MS/MS and Gene Ontology indicated that RSV positive NPS-EVs originate from different cellular sources, with the most abundant proteins from neutrophils and epithelial cells. A total of 490 piRNAs were detected by NGS sequencing of small RNA libraries obtained from NPS-EVs, which has not been reported prior to this study. Identification of inflammatory mediators and small non-coding RNAs which are compartmentalized in EVs contributes to understanding mechanisms of virus-mediated pathogenesis in RSV infections. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Graphical abstract

17 pages, 2540 KiB  
Review
Adaptor Protein Complexes in HIV-1 Pathogenesis: Mechanisms and Therapeutic Potential
by Maria Elena Barone, Alexis Lim, Madison Woody, Parisa Taklifi, Fatema Yeasmin, Kequan Wang, Mary K. Lewinski, Rajendra Singh, Charlotte A. Stoneham, Xiaofei Jia and John Guatelli
Viruses 2025, 17(5), 715; https://doi.org/10.3390/v17050715 - 16 May 2025
Viewed by 1964
Abstract
Adaptor protein (AP) complexes are critical components of the cellular membrane transport machinery. They mediate cargo selection during endocytosis and intracellular vesicular trafficking. Five AP complexes have been characterized (AP1-5), and together their roles extend to diverse cellular processes including the homeostasis of [...] Read more.
Adaptor protein (AP) complexes are critical components of the cellular membrane transport machinery. They mediate cargo selection during endocytosis and intracellular vesicular trafficking. Five AP complexes have been characterized (AP1-5), and together their roles extend to diverse cellular processes including the homeostasis of membranous organelles, membrane protein turnover, and immune responses. Human Immunodeficiency Virus type 1 (HIV-1) and other lentiviruses co-opt these complexes to support immune evasion and the assembly of maximally infectious particles. HIV-1 Nef interacts with AP1 and AP2 to manipulate intracellular trafficking and downregulate immune-related proteins such as CD4 and MHC-I. Vpu also co-opts AP1 and AP2, modulating the innate defense protein BST2 (Tetherin) and facilitating the release of virions from infected cells. The envelope glycoprotein (Env) hijacks AP complexes to reduce its expression at the cell surface and potentially support incorporation into virus particles. Some data suggest that Gag co-opts AP3 to drive assembly at intracellular compartments. In principle, targeting the molecular interfaces between HIV-1 proteins and AP complexes is a promising therapeutic approach. Blocking these interactions should impair HIV-1’s ability to produce infectious particles and evade immune defenses, leading to novel antivirals and facilitating a cure. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

19 pages, 1118 KiB  
Review
Long-Acting Extracellular Vesicle-Based Biologics in Osteoarthritis Immunotherapy
by Philip Drohat, Max Baron, Lee D. Kaplan, Thomas M. Best and Dimitrios Kouroupis
Bioengineering 2025, 12(5), 525; https://doi.org/10.3390/bioengineering12050525 - 15 May 2025
Cited by 1 | Viewed by 913
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by low-grade inflammation, cartilage breakdown, and persistent pain. Despite its prevalence, current therapeutic strategies primarily focus on symptom management rather than modifying disease progression. Monoclonal antibodies and cytokine inhibitors targeting inflammatory pathways, including TNF-α [...] Read more.
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by low-grade inflammation, cartilage breakdown, and persistent pain. Despite its prevalence, current therapeutic strategies primarily focus on symptom management rather than modifying disease progression. Monoclonal antibodies and cytokine inhibitors targeting inflammatory pathways, including TNF-α and IL-1, have shown promise but remain limited by inconsistent efficacy and safety concerns. Long-acting biologic therapies—ranging from extended-release formulations, such as monoclonal antibodies and cytokine inhibitors, to gene therapy approaches—have emerged as promising strategies to enhance treatment durability and improve patient outcomes. Extracellular vesicles (EVs) have gained particular attention as a novel delivery platform due to their inherent stability, biocompatibility, and ability to transport therapeutic cargo, including biologics and immunomodulatory agents, directly to joint tissues. This review explores the evolving role of EVs in OA treatment, highlighting their ability to extend drug half-life, improve targeting, and modulate inflammatory responses. Additionally, strategies for EV engineering, including endogenous and exogenous cargo loading, genetic modifications, and biomaterial-based delivery systems, are discussed. Full article
Show Figures

Figure 1

14 pages, 2998 KiB  
Review
The Potential Role of Exosomes in Communication Between Astrocytes and Endothelial Cells
by Joanna Czpakowska, Andrzej Głąbiński and Piotr Szpakowski
Int. J. Mol. Sci. 2025, 26(10), 4676; https://doi.org/10.3390/ijms26104676 - 14 May 2025
Cited by 1 | Viewed by 707
Abstract
Exosomes are extracellular vesicles secreted by almost all types of cells. Their release allows for the transport of specific regulatory cargo into the recipient cells and the modulation of their activity. Vesicular communication has also been identified as an important mechanism for the [...] Read more.
Exosomes are extracellular vesicles secreted by almost all types of cells. Their release allows for the transport of specific regulatory cargo into the recipient cells and the modulation of their activity. Vesicular communication has also been identified as an important mechanism for the regulation of numerous cellular activities in the brain tissue, contributing to proper neuronal functions and brain homeostasis. In this work, we focus on the role of exosomes and extracellular vesicles in the communication between astrocytes and brain endothelial cells, two major components of the blood–brain barrier. We perform a comprehensive review of the latest studies highlighting the role of exosomes in astrocyte-endothelial cell crosstalk within the blood–brain barrier. We have also described the role of particular exosomal miRNAs in the regulation of astrocytes and brain endothelial cell functions, and discuss some future implications. Full article
Show Figures

Figure 1

31 pages, 1806 KiB  
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 2 | Viewed by 1733
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

23 pages, 2304 KiB  
Review
Diagnostic and Therapeutic Utility of Extracellular Vesicles in Ocular Disease
by Vladimir Khristov, Sarah R. Weber, Mireille Caton-Darby, Gregory Campbell and Jeffrey M. Sundstrom
Int. J. Mol. Sci. 2025, 26(2), 836; https://doi.org/10.3390/ijms26020836 - 20 Jan 2025
Viewed by 1553
Abstract
Extracellular vesicles (EVs) are lipid bilayer particles released by virtually all cells, with prominent roles in both physiological and pathological processes. The size, number, and molecular composition of released EVs correlate to the cells of origin, modulated by the cell’s environment and pathologic [...] Read more.
Extracellular vesicles (EVs) are lipid bilayer particles released by virtually all cells, with prominent roles in both physiological and pathological processes. The size, number, and molecular composition of released EVs correlate to the cells of origin, modulated by the cell’s environment and pathologic state. The proteins, DNA, RNA, and protein cargo carried by EVs are protected by degradation, with a prominent role in targeted intercellular signaling. These properties make EVs salient targets as both carriers of biomarkers and potential therapeutic delivery vehicles. The majority of EV research has focused on blood, urine, saliva, and cerebrospinal fluid due to easy accessibility. EVs have also been identified and studied in all ocular biofluids, including the vitreous humor, the aqueous humor, and the tear film, and the study of EVs in ocular disease is a new, promising, and underexplored direction with unique challenges and considerations. This review covers recent advances in the diagnostic and therapeutic use of ocular EVs, with a focus on human applications and key preceding in vitro and in vivo animal studies. We also discuss future directions based on the study of EVs in other organ systems and disease sates. Full article
(This article belongs to the Special Issue The Role of Extracellular Vesicles in Cancers)
Show Figures

Figure 1

24 pages, 3089 KiB  
Review
Leveraging Microneedles for Raised Scar Management
by Zhengyun Jin, Young-Seong Kim and Joong Yeon Lim
Polymers 2025, 17(1), 108; https://doi.org/10.3390/polym17010108 - 2 Jan 2025
Cited by 2 | Viewed by 3403
Abstract
Disruption of the molecular pathways during physiological wound healing can lead to raised scar formation, characterized by rigid, thick scar tissue with associated symptoms of pain and pruritus. A key mechanical factor in raised scar development is excessive tension at the wound site. [...] Read more.
Disruption of the molecular pathways during physiological wound healing can lead to raised scar formation, characterized by rigid, thick scar tissue with associated symptoms of pain and pruritus. A key mechanical factor in raised scar development is excessive tension at the wound site. Recently, microneedles (MNs) have emerged as promising tools for scar management as they engage with scar tissue and provide them with mechanical off-loading from both internal and external sources. This review explores the mechanisms by which physical intervention of drug-free MNs alleviates mechanical tension on fibroblasts within scar tissue, thereby promoting tissue remodeling and reducing scar severity. Additionally, the role of MNs as an efficient cargo delivery system for the controlled and sustained release of a wide range of therapeutic agents into scar tissue is highlighted. By penetrating scar tissue, MNs facilitate controlled and sustained localized drug administration to modulate inflammation and fibroblastic cell growth. Finally, the remaining challenges and the future perspective of the field have been highlighted. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Figure 1

15 pages, 2167 KiB  
Article
Small Extracellular Vesicles Derived from Cord Blood Plasma and Placental Mesenchymal Stem Cells Attenuate Acute Lung Injury Induced by Lipopolysaccharide (LPS)
by Ranga P. Thiruvenkataramani, Amal Abdul-Hafez, Tulasi Kesaraju, Hend Mohamed, Sherif Abdelfattah Ibrahim, Amira Othman, Hattan Arif, Ahmed A. Zarea, Mohammed Abdulmageed, Myrna Gonzalez Arellano, Tarek Mohamed, Masamitsu Kanada, Burra V. Madhukar and Said A. Omar
Int. J. Mol. Sci. 2025, 26(1), 75; https://doi.org/10.3390/ijms26010075 - 25 Dec 2024
Cited by 2 | Viewed by 2398
Abstract
Sepsis is a risk factor associated with increasing neonatal morbidity and mortality, acute lung injury, and chronic lung disease. While stem cell therapy has shown promise in alleviating acute lung injury, its effects are primarily exerted through paracrine mechanisms rather than local engraftment. [...] Read more.
Sepsis is a risk factor associated with increasing neonatal morbidity and mortality, acute lung injury, and chronic lung disease. While stem cell therapy has shown promise in alleviating acute lung injury, its effects are primarily exerted through paracrine mechanisms rather than local engraftment. Accumulating evidence suggests that these paracrine effects are mediated by mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEVs), which play a critical role in immune system modulation and tissue regeneration. sEVs contain a diverse cargo of mRNA, miRNA, and proteins, contributing to their therapeutic potential. We hypothesize that sEVs derived from three distinct sources, cord blood plasma (CBP), Wharton jelly (WJ), and placental (PL) MSCs, may prevent the cytotoxicity induced by E. coli lipopolysaccharide (LPS) in lung alveolar epithelial cells. Objective: To determine the effects of CBP-, WJ-, and PL-MSCs-derived sEVs on cell viability, apoptosis, and proinflammatory cytokine production in alveolar epithelial cells and monocytes following LPS treatment. sEVs were collected from conditioned media of PL-MSCs, WJ-MSCs, and CBP using 50 nm membrane filters. sEVs were characterized based on nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), and Western blotting techniques. The protein concentration of isolated sEVs was used to standardize treatment doses. A549 cells and monocyte THP-1 cells were cultured and exposed to LPS in the presence or absence of sEVs for 72 h. Cell viability was measured using CellTiter-Glo 2.0 chemiluminescence-based assay. For cytokine analysis, A549 and THP-1 cells were pre-incubated for 24 h with or without PL- and CBP-sEVs, followed by exposure to LPS or control conditions for an additional 24 h. The conditioned media were collected, and interleukin-6 (IL-6) and interleukin-8 (IL-8) levels were quantified using ELISA. LPS treatment significantly reduced the viability of both A549 and THP-1 cells. The presence of CB- or WJ-sEVs significantly increased cell viability compared to controls. Cells treated with PL-sEVs showed increased cell viability but did not reach statistical significance. LPS-treated cells showed a significant increase in apoptosis and elevated levels of pro-inflammatory cytokines IL-6 and IL-8. All three sEVs types (CBP-, WJ-, and PL-sEVs) significantly reduced LPS-induced apoptosis and IL-6 release. Interestingly, while WJ-sEVs decreased IL-8, both CBP- and PL-sEVs led to an increase in IL-8 compared to their respective controls. CBP-, PL-, and WJ-derived sEVs demonstrated protective effects against LPS-induced injury in alveolar epithelial cells and monocytes, as evidenced by increased cell viability and modulation of pro-inflammatory cytokine release. These findings suggest that placenta-derived sEVs have the potential to modulate the immune response, mitigate inflammation, and prevent end-organ damage in neonatal sepsis. Full article
(This article belongs to the Special Issue Exosomes—3rd Edition)
Show Figures

Figure 1

37 pages, 4357 KiB  
Article
Low Magnetic Field Exposure Alters Prostate Cancer Cell Properties
by Sigrun Lange, Jameel M. Inal, Igor Kraev, Dafydd Alwyn Dart and Pinar Uysal-Onganer
Biology 2024, 13(9), 734; https://doi.org/10.3390/biology13090734 - 19 Sep 2024
Cited by 1 | Viewed by 2994
Abstract
Prostate cancer is the second most common neoplasia and fifth-leading cause of cancer death in men worldwide. Electromagnetic and magnetic fields have been classified as possible human carcinogens, but current understanding of molecular and cellular pathways involved is very limited. Effects due to [...] Read more.
Prostate cancer is the second most common neoplasia and fifth-leading cause of cancer death in men worldwide. Electromagnetic and magnetic fields have been classified as possible human carcinogens, but current understanding of molecular and cellular pathways involved is very limited. Effects due to extremely low magnetic/hypomagnetic fields (LMF) are furthermore poorly understood. Extracellular vesicles (EVs) are crucial mediators of cellular communication with multifaceted roles in cancer progression, including via transport and uptake of various protein and microRNA (miRNA) EV-cargoes. miRNAs regulate gene expression and are implicated in cancer-related processes such as proliferation, metastasis, and chemoresistance. This study investigated the effects of LMF exposure (20 nT) by magnetic shielding on the prostate cancer cell line PC3 compared to the prostate epithelial cell line PNT2 under short-term (4 h) conditions. We examined EV profiles following a 4 h LMF exposure alongside associated functional enrichment KEGG and GO pathways for the EV proteomes. The 4 h LMF exposure significantly reduced cellular EV release and modified PC3 EV cargoes to a more inflammatory and metastatic profile, with 16 Disease Pathways and 95 Human Phenotypes associated specifically with the LMF-treated PC3 EV proteomes. These included cancerous, metabolic, blood, skin, cardiac and skeletal Disease Pathways, as well as pain and developmental disorders. In the normal PNT2 cells, less EV protein cargo was observed following LMF exposure compared with cells not exposed to LMF, and fewer associated functional enrichment pathways were identified. This pointed to some differences in various cellular functions, ageing, defence responses, oxidative stress, and disease phenotypes, including respiratory, digestive, immune, and developmental pathways. Furthermore, we analysed alterations in matrix metalloproteinases (MMPs) and miRNAs linked to metastasis, as this is crucial in cancer aggressiveness. The 4 h LMF exposure caused a significant increase in MMP2 and MMP9, as well as in onco-miRs miR-155, miR-210, miR-21, but a significant reduction in tumour-suppressor miRs (miR-200c and miR-126) in the metastatic PC3 cells, compared with normal PNT2 cells. In addition, 4 h LMF exposure significantly induced cellular invasion of PC3 cells. Overall, our findings suggest that changes in magnetic field exposures modulate EV-mediated and miR-regulatory processes in PCa metastasis, providing a basis for exploring novel therapeutic strategies. Full article
(This article belongs to the Special Issue The Rules of Life Rethought: Latest Progress in Quantum Biology)
Show Figures

Figure 1

17 pages, 2828 KiB  
Article
Proteomic Profile of Circulating Extracellular Vesicles in the Brain after Δ9-Tetrahydrocannabinol Inhalation
by Valeria Lallai, TuKiet T. Lam, Rolando Garcia-Milian, Yen-Chu Chen, James P. Fowler, Letizia Manca, Daniele Piomelli, Kenneth Williams, Angus C. Nairn and Christie D. Fowler
Biomolecules 2024, 14(9), 1143; https://doi.org/10.3390/biom14091143 - 10 Sep 2024
Viewed by 1716
Abstract
Given the increasing use of cannabis in the US, there is an urgent need to better understand the drug’s effects on central signaling mechanisms. Extracellular vesicles (EVs) have been identified as intercellular signaling mediators that contain a variety of cargo, including proteins. Here, [...] Read more.
Given the increasing use of cannabis in the US, there is an urgent need to better understand the drug’s effects on central signaling mechanisms. Extracellular vesicles (EVs) have been identified as intercellular signaling mediators that contain a variety of cargo, including proteins. Here, we examined whether the main psychoactive component in cannabis, Δ9-tetrahydrocannabinol (THC), alters EV protein signaling dynamics in the brain. We first conducted in vitro studies, which found that THC activates signaling in choroid plexus epithelial cells, resulting in transcriptional upregulation of the cannabinoid 1 receptor and immediate early gene c-fos, in addition to the release of EVs containing RNA cargo. Next, male and female rats were examined for the effects of either acute or chronic exposure to aerosolized (‘vaped’) THC on circulating brain EVs. Cerebrospinal fluid was extracted from the brain, and EVs were isolated and processed with label-free quantitative proteomic analyses via high-resolution tandem mass spectrometry. Interestingly, circulating EV-localized proteins were differentially expressed based on acute or chronic THC exposure in a sex-specific manner. Taken together, these findings reveal that THC acts in the brain to modulate circulating EV signaling, thereby providing a novel understanding of how exogenous factors can regulate intercellular communication in the brain. Full article
(This article belongs to the Special Issue Advances in Neuroproteomics)
Show Figures

Figure 1

23 pages, 3714 KiB  
Article
The Six-Transmembrane Enzyme GDE2 Is Required for the Release of Molecularly Distinct Small Extracellular Vesicles from Neurons
by Kyle T. Shuler, Josue Llamas-Rodriguez, Reuben Levy-Myers and Shanthini Sockanathan
Cells 2024, 13(17), 1414; https://doi.org/10.3390/cells13171414 - 24 Aug 2024
Cited by 1 | Viewed by 1475
Abstract
Extracellular vesicles (EVs) are implicated in a multitude of physiological and pathophysiological processes in the nervous system; however, their biogenesis and cargoes are not well defined. Glycerophosphodiester Phosphodiesterase 2 (GDE2 or GDPD5) is a six-transmembrane protein that cleaves the Glycosylphosphatidylinositol (GPI)-anchor that tethers [...] Read more.
Extracellular vesicles (EVs) are implicated in a multitude of physiological and pathophysiological processes in the nervous system; however, their biogenesis and cargoes are not well defined. Glycerophosphodiester Phosphodiesterase 2 (GDE2 or GDPD5) is a six-transmembrane protein that cleaves the Glycosylphosphatidylinositol (GPI)-anchor that tethers some proteins to the membrane and has important roles in neurodevelopment and disease-relevant pathways of neuronal survival. We show here that GDE2 regulates the number of small EVs (sEVs) released from the cell surface of neurons via its GPI-anchor cleavage activity and contributes to the loading of protein cargo through enzymatic and non-enzymatic mechanisms. Proteomic profiling reveals that GDE2 releases at least two distinct EV populations, one containing GDE2 itself and the other harboring the putative ectosomal markers CD9 and BSG. sEVs released by GDE2 are enriched in cytoskeletal and actin-remodeling proteins, suggesting a potential mechanism for GDE2-dependent EV release. Further, sEV populations released by GDE2 are enriched in proteins responsible for modulating synaptic activity and proteins that are critical for cellular redox homeostasis. These studies identify GDE2 as a novel regulator of molecularly distinct sEV populations from neurons with potential roles in the synaptic and redox pathways required for neuronal function and survival. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

21 pages, 1556 KiB  
Review
Oncoviral Infections and Small Extracellular Vesicles
by Łukasz Ważny, Theresa L. Whiteside and Monika Pietrowska
Viruses 2024, 16(8), 1291; https://doi.org/10.3390/v16081291 - 13 Aug 2024
Cited by 2 | Viewed by 2322
Abstract
Small extracellular vesicles (sEV) are small membrane-bound nanovesicles with a size range below 200 nm that are released by all types of cells. sEV carry a diverse cargo of proteins, lipids, glycans, and nucleic acids that mimic the content of producer cells. sEV [...] Read more.
Small extracellular vesicles (sEV) are small membrane-bound nanovesicles with a size range below 200 nm that are released by all types of cells. sEV carry a diverse cargo of proteins, lipids, glycans, and nucleic acids that mimic the content of producer cells. sEV mediate intercellular communication and play a key role in a broad variety of physiological and pathological conditions. Recently, numerous reports have emerged examining the role of sEV in viral infections. A significant number of similarities in the sEV biogenesis pathways and the replication cycles of viruses suggest that sEV might influence the course of viral infections in diverse ways. Besides directly modulating virus propagation by transporting the viral cargo (complete virions, proteins, RNA, and DNA), sEV can also modify the host antiviral response and increase the susceptibility of cells to infection. The network of mutual interactions is particularly complex in the case of oncogenic viruses, deserving special consideration because of its significance in cancer progression. This review summarizes the current knowledge of interactions between sEV and oncogenic viruses, focusing on sEV abilities to modulate the carcinogenic properties of oncoviruses. Full article
(This article belongs to the Special Issue Molecular and Cellular Biology of Human Oncogenic Viruses)
Show Figures

Figure 1

23 pages, 1484 KiB  
Review
Stress-Related Roles of Exosomes and Exosomal miRNAs in Common Neuropsychiatric Disorders
by Myrsini Chamakioti, George P. Chrousos, Eva Kassi, Dimitrios Vlachakis and Christos Yapijakis
Int. J. Mol. Sci. 2024, 25(15), 8256; https://doi.org/10.3390/ijms25158256 - 29 Jul 2024
Cited by 4 | Viewed by 2531
Abstract
Exosomes, natural nanovesicles that contain a cargo of biologically active molecules such as lipids, proteins, and nucleic acids, are released from cells to the extracellular environment. They then act as autocrine, paracrine, or endocrine mediators of communication between cells by delivering their cargo [...] Read more.
Exosomes, natural nanovesicles that contain a cargo of biologically active molecules such as lipids, proteins, and nucleic acids, are released from cells to the extracellular environment. They then act as autocrine, paracrine, or endocrine mediators of communication between cells by delivering their cargo into recipient cells and causing downstream effects. Exosomes are greatly enriched in miRNAs, which are small non-coding RNAs that act both as cytoplasmic post-transcriptional repression agents, modulating the translation of mRNAs into proteins, as well as nuclear transcriptional gene activators. Neuronal exosomal miRNAs have important physiologic functions in the central nervous system (CNS), including cell-to-cell communication, synaptic plasticity, and neurogenesis, as well as modulating stress and inflammatory responses. Stress-induced changes in exosomal functions include effects on neurogenesis and neuroinflammation, which can lead to the appearance of various neuropsychiatric disorders such as schizophrenia, major depression, bipolar disorder, and Alzheimer’s and Huntington’s diseases. The current knowledge regarding the roles of exosomes in the pathophysiology of common mental disorders is discussed in this review. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

15 pages, 313 KiB  
Review
Progress on the Regulation of the Host Immune Response by Parasite-Derived Exosomes
by Xinyue Zhang, Chuanxin Yu and Lijun Song
Pathogens 2024, 13(8), 623; https://doi.org/10.3390/pathogens13080623 - 26 Jul 2024
Cited by 2 | Viewed by 2651
Abstract
Exosomes are membrane-bound structures released by cells into the external environment that carry a significant amount of important cargo, such as proteins, DNA, RNA, and lipids. They play a crucial role in intercellular communication. Parasites have complex life cycles and can release exosomes [...] Read more.
Exosomes are membrane-bound structures released by cells into the external environment that carry a significant amount of important cargo, such as proteins, DNA, RNA, and lipids. They play a crucial role in intercellular communication. Parasites have complex life cycles and can release exosomes at different stages. Exosomes released by parasitic pathogens or infected cells contain parasitic nucleic acids, antigenic molecules, virulence factors, drug-resistant proteins, proteases, lipids, etc. These components can regulate host gene expression across species or modulate signaling pathways, thereby dampening or activating host immune responses, causing pathological damage, and participating in disease progression. This review focuses on the means by which parasitic exosomes modulate host immune responses, elaborates on the pathogenic mechanisms of parasites, clarifies the interactions between parasites and hosts, and provides a theoretical basis and research directions for the prevention and treatment of parasitic diseases. Full article
(This article belongs to the Special Issue Advances in Host-Helminth Interactions)
Show Figures

Graphical abstract

Back to TopTop