Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.822
Filter
1.
Biomaterials ; 313: 122804, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39236631

ABSTRACT

Insulin resistance and pancreatic ß-cell dysfunction are the main pathogenesis of type 2 diabetes mellitus (T2DM). However, insulin therapy and diabetes medications do not effectively solve the two problems simultaneously. In this study, a biomimetic oral hydrogen nanogenerator that leverages the benefits of edible plant-derived exosomes and hydrogen therapy was constructed to overcome this dilemma by modulating gut microbiota and ameliorating oxidative stress and inflammatory responses. Hollow mesoporous silica (HMS) nanoparticles encapsulating ammonia borane (A) were used to overcome the inefficiency of H2 delivery in traditional hydrogen therapy, and exosomes originating from ginger (GE) were employed to enhance biocompatibility and regulate intestinal flora. Our study showed that HMS/A@GE not only considerably ameliorated insulin resistance and liver steatosis, but inhibited the dedifferentiation of islet ß-cell and enhanced pancreatic ß-cell proportion in T2DM model mice. In addition to its antioxidant and anti-inflammatory effects, HMS/A@GE augmented the abundance of Lactobacilli spp. and tryptophan metabolites, such as indole and indole acetic acid, which further activated the AhR/IL-22 pathway to improve intestinal-barrier function and metabolic impairments. This study offers a potentially viable strategy for addressing the current limitations of diabetes treatment by integrating gut-microbiota remodelling with antioxidant therapies.


Subject(s)
Antioxidants , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Insulin Resistance , Insulin-Secreting Cells , Nanoparticles , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Antioxidants/pharmacology , Gastrointestinal Microbiome/drug effects , Nanoparticles/chemistry , Mice , Male , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Inbred C57BL , Zingiber officinale/chemistry , Silicon Dioxide/chemistry , Exosomes/metabolism , Biomimetics/methods , Oxidative Stress/drug effects
2.
Results Probl Cell Differ ; 73: 249-297, 2024.
Article in English | MEDLINE | ID: mdl-39242383

ABSTRACT

The landscape of exosome research has undergone a significant paradigm shift, with a departure from early conceptions of exosomes as vehicles for cellular waste disposal towards their recognition as integral components of cellular communication with therapeutic potential. This chapter presents an exhaustive elucidation of exosome biology, detailing the processes of exosome biogenesis, release, and uptake, and their pivotal roles in signal transduction, tissue repair, regeneration, and intercellular communication. Additionally, the chapter highlights recent innovations and anticipates future directions in exosome research, emphasizing their applicability in clinical settings. Exosomes have the unique ability to navigate through tissue spaces to enter the circulatory system, positioning them as key players in tissue repair. Their contributory role in various processes of tissue repair, although in the nascent stages of investigation, stands out as a promising area of research. These vesicles function as a complex signaling network for intracellular and organ-level communication, critical in both pathological and physiological contexts. The chapter further explores the tissue-specific functionality of exosomes and underscores the advancements in methodologies for their isolation and purification, which have been instrumental in expanding the scope of exosome research. The differential cargo profiles of exosomes, dependent on their cellular origin, position them as prospective diagnostic biomarkers for tissue damage and regenerative processes. Looking ahead, the trajectory of exosome research is anticipated to bring transformative changes to biomedical fields. This includes advancing diagnostic and prognostic techniques that utilize exosomes as non-invasive biomarkers for a plethora of diseases, such as cancer, neurodegenerative, and cardiovascular conditions. Additionally, engineering exosomes through alterations of their native content or surface properties presents a novel frontier, including the synthesis of artificial or hybrid variants with enhanced functional properties. Concurrently, the ethical and regulatory frameworks surrounding exosome research, particularly in clinical translation, will require thorough deliberation. In conclusion, the diverse aspects of exosome research are coalescing to redefine the frontiers of diagnostic and therapeutic methodologies, cementing its importance as a discipline of considerable consequence in the biomedical sciences.


Subject(s)
Cell Communication , Exosomes , Exosomes/metabolism , Humans , Cell Communication/physiology , Animals , Wound Healing/physiology , Signal Transduction/physiology
3.
Results Probl Cell Differ ; 73: 301-326, 2024.
Article in English | MEDLINE | ID: mdl-39242384

ABSTRACT

Cell-to-cell interactions are essential for proper development, homeostasis, and complex syncytia/organ formation and function. Intercellular communication are mediated by multiple mechanisms including soluble mediators, adhesion molecules and specific mechanisms of cell to cell communication such as Gap junctions (GJ), tunneling nanotubes (TNT), and exosomes. Only recently, has been discovered that TNTs and exosomes enable the exchange of large signaling molecules, RNA, viral products, antigens, and organelles opening new avenues of research and therapeutic approaches. The focus of this review is to summarize these recent findings in physiologic and pathologic conditions.


Subject(s)
Cell Communication , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/metabolism , Cell Communication/physiology , Animals , Gap Junctions/metabolism , Exosomes/metabolism
4.
Sci Rep ; 14(1): 21669, 2024 09 17.
Article in English | MEDLINE | ID: mdl-39289425

ABSTRACT

A novel core-shell nanocarrier system has been designed for co-delivery of a small anticancer drug, docetaxel (DTX) and tumor suppressor (TS) miR-34a named as Exo(PAN34a+DTX). The core is formed by pH dependent polyamine salt aggregates (PSA) containing both the payloads and the shell is formed by RAW 264.7 cell derived exosomal fragments. Herein, phosphate driven polyallylamine hydrochloride (PAH, MW:17,500 Da) PSA was formed in presence of miR-34a and DTX to form PAN34a+DTX. The formulation exhibited pH dependent DTX release with only 33.55 ± 2.12% DTX release at pH 7.2 and 75.21 ± 1.8% DTX release till 144 h at pH 5.5. At 1.21 molar ratio of phosphate to the amine (known as R value), efficient complexation of miR-34a (3.6 µM) in the PAN particles was obtained. PAN34a+DTX demonstrated particle size (163.86 ± 12.89 nm) and zeta-potential value of 17.53 ± 5.10 mV which upon exosomal fragment layering changed to - 7.23 ± 2.75 mV which is similar to the zeta-potential of the exosomal fragments, i.e., - 8.40 ± 1.79 mV. The final formulation Exo(PAN34a+DTX), loaded with 40 ng/mL DTX and 50 nM miR-34a exhibited 48.20 ± 4.59% cytotoxicity in triple negative breast cancer (TNBC) cells, 4T1. Co-localization of CM-DiI (red fluorescence) stained exosomal fragments and FAM-siRNA (green fluorescence) in the cytoplasm of 4T1 cells after 6 h of Exo(PANFAM) treatment confirmed the efficiency of the designed system to co-deliver two actives. Exo(PAN34a+DTX) also reduced BCL-2 expression (target gene for miR-34a) by 8.98 folds in comparison to free DTX confirming promising co-delivery and apoptosis inducing effect of Exo(PAN34a+DTX) in 4T1.


Subject(s)
Apoptosis , Docetaxel , Exosomes , MicroRNAs , Polyamines , MicroRNAs/genetics , MicroRNAs/metabolism , Docetaxel/pharmacology , Docetaxel/administration & dosage , Polyamines/chemistry , Humans , Exosomes/metabolism , Apoptosis/drug effects , Animals , Mice , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , RAW 264.7 Cells , Cell Line, Tumor , Drug Carriers/chemistry
5.
Zhonghua Fu Chan Ke Za Zhi ; 59(9): 710-718, 2024 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-39313423

ABSTRACT

Objective: To investigate the impact of exosomes and microRNA (miRNA) from placental mesenchymal stem cells on chemotherapy-damaged ovarian granulosa cells. Methods: Various public databases were searched for miRNA targeting phosphatase and tensin homologue deleted on chromosome 10 (PTEN) gene. After miRNA transfection into human ovarian granulosa cells, cell growth and expressions of the target miRNA and PTEN were detected. Cisplatin was utilized to induce damage to human ovarian granulosa cells, which were subsequently co-cultured with human placental mesenchymal stem cells and exosomes generated from mesenchymal stem cells, then apoptosis and expressions of PTEN and the target miRNA were detected. Results: After analyzing several databases, miRNA 139-5p (miR-139-5p) was chosen as the target miRNA for this research. Transfection of miR-139-5p mimics into human ovarian granulosa cells elevated miR-139-5p expression level (9 882.080±1 049.130), reduced PTEN protein expression level (0.78±0.11), and increased cell proliferation absorbance (0.85±0.07). Cisplatin treatment severely damaged human ovarian granulosa cells and increased apoptosis, cisplatin-treated cells had a higher apoptosis ratio compared to untreated cells [ (41.9±1.0)% vs (5.0±0.3)%, P<0.001]. In damaged human ovarian granulosa cells, co-cultured with human placental mesenchymal stem cells and exosomes increased miR-139-5p expression levels (1.31±0.04 and 1.20±0.03, respectively) and decreased apoptosis ratios [(20.0±0.4)% and (22.3±1.1)%, respectively]. Conclusion: Placental mesenchymal stem cell-derived exosomes repair damages of cisplatin-induced ovarian granulosa cell and could target PTEN gene through miR-139-5p, which might be a potential option for the treatment of chemotherapy-induced ovarian dysfunction.


Subject(s)
Apoptosis , Cell Proliferation , Cisplatin , Exosomes , Granulosa Cells , Mesenchymal Stem Cells , MicroRNAs , PTEN Phosphohydrolase , Placenta , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Exosomes/metabolism , Exosomes/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Granulosa Cells/metabolism , Cisplatin/adverse effects , Placenta/metabolism , Pregnancy , Transfection , Coculture Techniques , Ovary , Antineoplastic Agents/adverse effects
6.
Mol Biol Rep ; 51(1): 995, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39298063

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts, as a major component of the tumor microenvironment, have been shown to exhibit protumorigenic effects in pancreatic ductal adenocarcinoma. Moreover, cancer-associated fibroblasts-derived exosomes have been reported to promote tumor development, but exact mechanisms have not been elucidated. The purpose of this study was to investigate the processes by which exosomes generated from cancer-associated fibroblasts promote tumor growth. METHODS: twenty-one patients with pancreatic ductal adenocarcinoma who evaluated preoperatively as potentially surgically resectable without distant metastasis and pathologically examined postoperatively as pancreatic ductal cell carcinoma were included. We determined the expression of Leptin as well as downstream proteins at the clinical and cellular levels. Cancer-associated fibroblast-derived exosomes were characterised by nanoparticle transmission electron microscopy and tracking analysis. To ascertain the mechanism mediating the action of exosomal Leptin in pancreatic ductal adenocarcinoma, we performed CCK-8 assay, colony formation assays, transwell and wound healing assays in PSN1 cells to evaluate cell proliferation, migration and invasion. Western blotting was used to detect the level of Leptin, ABL2 and exosome markers. qRT-PCR was employed to evaluate miR-224-3p. Cancer-associated fibroblasts markers and exosome uptake were verified by immunofluorescence. RESULTS: Western blotting assays show that Leptin is present inside tissues and cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Cancer-associated fibroblasts stimulated PSN1 cells growth, migration and invasion in vitro by secreting the exosomal Leptin. Exosomal Leptin could regulate miR-224-3p, which targets negative regulation of ABL2. Inhibiting Leptin significantly limited PSN1 cells growth, migration and invasion. In vitro analyses revealed that miR-224-3p mimics mitigate the inhibitory effect of cancer-associated fibroblasts knockdown of Leptin on PSN1 cells development, but overexpression of ABL2 partly abolished the tumor-promoting phenotype of miR-224-3p mimics. CONCLUSION: Our results revealed that cancer-associated fibroblasts mediate pancreatic ductal adenocarcinoma development by regulating the miR-224-3p/ABL2 molecular axis through the secretion of the exosomal Leptin.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Cell Movement , Cell Proliferation , Exosomes , Gene Expression Regulation, Neoplastic , Leptin , MicroRNAs , Pancreatic Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/metabolism , Exosomes/genetics , Leptin/metabolism , Leptin/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cancer-Associated Fibroblasts/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Male , Female , Middle Aged , Cell Line, Tumor , Cell Proliferation/genetics , Cell Movement/genetics , Aged , Tumor Microenvironment/genetics
7.
FASEB J ; 38(18): e70062, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39305125

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with impaired adipose tissue physiology. Elevated brown adipose tissue (BAT) mass or activity has shown potential in the treatment of PCOS. In this study, we aimed to investigate whether BAT-derived exosomes (BAT-Exos), as potential biomarkers of BAT activity, exert similar benefits as BAT in the treatment of PCOS. PCOS was induced in female C57BL/6J mice orally administered 1 mg/kg of letrozole for 21 days. Subsequently, the animals underwent transplantation with BAT or administered BAT-Exos (200 µg) isolated from young healthy mice via the tail vein; healthy female mice were used as controls. The results indicate that BAT-Exos treatment significantly reduced body weight and improved insulin resistance in PCOS mice. In addition, BAT-Exos improved ovulation function by reversing the acyclicity of the estrous cycle, decreasing circulating luteinizing hormone and testosterone, recovering ovarian performance, and improving oocyte quality, leading to a higher pregnancy rate and litter size. Furthermore, western blotting revealed reduced expression of signal transducer and activator of transcription 3 (STAT3) and increased expression of glutathione peroxidase 4 (GPX4) in the ovaries of mice in the BAT-Exos group. To further explore the role of the STAT3/GPX4 signaling pathway in PCOS mice, we treated the mice with an intraperitoneal injection of 5 mg/kg stattic, a STAT3 inhibitor. Consistent with BAT-Exos treatment, the administration of stattic rescued letrozole-induced PCOS phenotypes. These findings suggest that BAT-Exos treatment might be a potential therapeutic strategy for PCOS and that the STAT3/GPX4 signaling pathway is a critical therapeutic target for PCOS.


Subject(s)
Adipose Tissue, Brown , Exosomes , Mice, Inbred C57BL , Phospholipid Hydroperoxide Glutathione Peroxidase , Polycystic Ovary Syndrome , STAT3 Transcription Factor , Signal Transduction , Animals , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/therapy , Female , Mice , STAT3 Transcription Factor/metabolism , Exosomes/metabolism , Adipose Tissue, Brown/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Letrozole/pharmacology , Insulin Resistance , Ovary/metabolism
8.
Nutr Diabetes ; 14(1): 75, 2024 09 13.
Article in English | MEDLINE | ID: mdl-39271650

ABSTRACT

OBJECTIVE: The release of adipose tissue-derived miRNAs is increased under conditions of obesity, but the exact molecular mechanisms involved have not been elucidated. This study investigated whether obesity-induced increases in palmitic acid (PA) content could activate the NF-κB/endoplasmic reticulum stress (ER stress) pathway and promote the expression and release of exosomal miRNAs in adipocytes. METHODS: Abdominal adipose tissue and serum samples were collected from normal weight individuals and people with obesity to clarify the correlation of serum PA content with NF-κB/ER stress and the release of exosomal miRNAs. NF-κB and ER stress were blocked in obese mice and in vitro cultured adipocytes to demonstrate the molecular mechanisms by which PA promotes the release of exosomal miRNAs.The morphology, particle size and distribution of the exosomes were observed via transmission electron microscopy and NTA. RESULTS: Accompanied by increased serum PA levels, the NF-κB/ER stress pathway was activated in the adipose tissue of people with obesity and in high-fat diet (HFD)-induced obese mice; moreover, the levels of miRNAs in both adipose tissue and serum were increased. P-p65 (Bay11-7082) and ER stress (TUDCA) blockers significantly reduced the levels of miRNAs in abdominal adipose tissue and serum, decreased blood glucose levels, and improved glucose tolerance and insulin sensitivity in obese mice. In 3T3-L1 adipocytes, high concentrations of PA activated the NF-κB/ER stress pathway and increased the expression and release of miRNAs in exosomes. P-p65 (Bay11-7082) and ER stress (TUDCA) blockers significantly reversed the increased release exosomal miRNAs cause by PA. CONCLUSIONS: Obesity-induced increases in PA content increase the expression and release of miRNAs in adipocyte exosomes by activating the NF-κB/ER stress pathway.


Subject(s)
Adipocytes , Diet, High-Fat , Endoplasmic Reticulum Stress , Obesity , Palmitic Acid , Signal Transduction , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , 3T3-L1 Cells , Abdominal Fat/metabolism , Adipocytes/metabolism , Exosomes/metabolism , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/metabolism , NF-kappa B/metabolism , Obesity/metabolism , Palmitic Acid/pharmacology
9.
Bull Exp Biol Med ; 177(4): 544-551, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39279005

ABSTRACT

We developed a model of inflammation and airway remodeling in C57 mice provoked by exosomes derived from bone marrow mesenchymal stem cells infected by respiratory syncytial virus (RSV). The mean size of control and infected exosomes in vitro were 167.9 and 118.5 nm, respectively. After induction of modeled pathology, the severity of airway inflammation and its remodeling were analyzed by histopathological methods. In addition, the blood levels of inflammatory factors IL-10, IL-17, transforming growth factor-ß (TGF-ß), and TNFα were assayed; in the lung tissues, the expression levels of MMP-2, MMP-9, α-smooth muscle actin (α-SMA), and TGF-ß were measured. In the developed model, the effects of RSV-induced and non-induced exosomes were compared with those of inactivated and non-inactivated RSV. Intranasal administration of RSV-induced exosomes decreased the levels of serum inflammatory factors IL-10 and IL-17 and increased the expression of serum proinflammatory cytokine TNFα. Increased levels of MMP-2, MMP-9, and α-SMA, enhanced expression of TGF-ß in the lung tissue, and pathological staining of the lung tissues indicated infiltration with inflammatory cells and luminal constriction. Thus, RSV-induced exosomes can provoke airway inflammation and remodeling in mice similar to RSV, while non-induced exosomes cannot produce such alterations.


Subject(s)
Airway Remodeling , Disease Models, Animal , Exosomes , Interleukin-10 , Interleukin-17 , Matrix Metalloproteinase 2 , Mesenchymal Stem Cells , Mice, Inbred C57BL , Respiratory Syncytial Virus Infections , Tumor Necrosis Factor-alpha , Animals , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Matrix Metalloproteinase 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Interleukin-10/metabolism , Interleukin-10/blood , Interleukin-17/metabolism , Lung/pathology , Lung/virology , Lung/metabolism , Matrix Metalloproteinase 9/metabolism , Respiratory Syncytial Viruses/pathogenicity , Transforming Growth Factor beta/metabolism , Actins/metabolism , Inflammation/pathology , Inflammation/metabolism , Bone Marrow Cells/metabolism , Female
10.
BMC Cancer ; 24(1): 1173, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304856

ABSTRACT

BACKGROUND: Exosomes are closely associated with different aspects of tumor-progression in patients with head and neck squamous cell carcinoma (HNSCC), such as angiogenesis or immune regulation. As extracellular vesicles they are involved in the intercellular communication by transferring their cargo such as proteins and nucleic acids from one cell to another. However, the influence of tumor related plasma-derived exosomes on the polarization and characteristics of monocyte derived macrophages is not fully understood. METHODS: Exosomes were isolated from plasma samples of healthy donors (HD) and HNSCC patients and further evaluated with regard to morphology, size and protein composition via transmission electron microscopy, nanoparticle tracking, western blot analysis and cytokine assays. Differentiation and characteristics of monocyte derived macrophages upon exosome internalization were analyzed using flow cytometry and fluorescence microscopy. Macrophage cytokine secretion patterns were analyzed by human cytokine antibody arrays and ELISA measurements. RESULTS: Our data revealed elevated overall plasma levels of CTLA-4, PD-L1, and TIM-3 as well as elevated exosome-associated CTLA-4, PD-L2, TIM-3, and LAG-3 levels in HNSCC patients compared to HD. Furthermore, we observed a significant type 2-like polarization and elevated CXCL4 secretion of monocyte derived macrophages upon internalization of plasma-derived exosomes from HNSCC patients, which could be visualized by fluorescence microcopy of membrane stained exosomes. CONCLUSIONS: The study provides new insights regarding exosome driven pro-tumorigenic immune regulation in the circulation of patients with head and neck cancer and could help to better understand the individual immunologic situation.


Subject(s)
Exosomes , Head and Neck Neoplasms , Macrophages , Humans , Exosomes/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/blood , Macrophages/metabolism , Macrophages/immunology , Male , Female , Platelet Factor 4/metabolism , Middle Aged , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/blood , Aged , Adult
11.
J Agric Food Chem ; 72(38): 21030-21040, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39283309

ABSTRACT

Mammalian milk exosomal miRNAs play an important role in maintaining intestinal immune homeostasis and protecting epithelial barrier function, but the specific miRNAs and whether miRNA-mediated mechanisms are responsible for these benefits remain a matter of investigation. This study isolated sheep milk-derived exosomes (sheep MDEs), identifying the enriched miRNAs in sheep MDEs, oar-miR-148a, and oar-let-7b as key components targeting TLR4 and TRAF1, which was validated by a dual-luciferase reporter assay. In dextran sulfate sodium-induced colitis mice, administration of sheep MDEs alleviated colitis symptoms, reduced colonic inflammation, and systemic oxidative stress, as well as significantly increased colonic oar-miR-148a and oar-let-7b while reducing toll-like receptor 4 (TLR4) and TNF-receptor-associated factor 1 (TRAF1) level. Further characterization in TNF-α-challenged Caco-2 cells showed that overexpression of these miRNAs suppressed the TLR4/TRAF1-IκBα-p65 pathway and reduced IL-6 and IL-12 production. These findings indicate that sheep MDEs exert gastrointestinal anti-inflammatory effects through the miRNA-mediated modulation of TLR4 and TRAF1, highlighting their potential in managing colitis.


Subject(s)
Colitis , Dextran Sulfate , Exosomes , MicroRNAs , Milk , TNF Receptor-Associated Factor 1 , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/immunology , Dextran Sulfate/adverse effects , Milk/chemistry , Milk/metabolism , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Mice , Sheep , Humans , Exosomes/genetics , Exosomes/metabolism , Exosomes/chemistry , Exosomes/immunology , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/metabolism , Caco-2 Cells , Male , Mice, Inbred C57BL , Female
12.
Lab Chip ; 24(19): 4581-4593, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39230477

ABSTRACT

The gut communicates with the brain in a variety of ways known as the gut-brain axis (GBA), which is known to affect neurophysiological functions as well as neuronal disorders. Exosomes capable of passing through the blood-brain-barrier (BBB) have received attention as a mediator of gut-brain signaling and drug delivery vehicles. In conventional well plate-based experiments, it is difficult to observe the exosome movement in real time. Here, we developed a microfluidic-based GBA chip for co-culturing gut epithelial cells and neuronal cells and simultaneously observing exosome transport. The GBA-chip is aimed to mimic the in vivo situation of convective flow in blood vessels and convective and diffusive transport in the tissue interstitium. Here, fluorescence-labeled exosome was produced by transfection of HEK-293T cells with CD63-GFP plasmid. We observed in real time the secretion of CD63-GFP-exosomes by the transfected HEK-293T cells in the chip, and transport of the exosomes to neuronal cells and analyzed the dynamics of GFP-exosome movement. Our model is expected to enhance understanding of the roles of exosome in GBA.


Subject(s)
Brain , Exosomes , Lab-On-A-Chip Devices , Humans , Exosomes/metabolism , HEK293 Cells , Brain/metabolism , Brain/cytology , Coculture Techniques/instrumentation , Microfluidic Analytical Techniques/instrumentation , Neurons/metabolism , Neurons/cytology , Biological Transport , Blood-Brain Barrier/metabolism , Models, Biological
13.
Theranostics ; 14(14): 5698-5724, 2024.
Article in English | MEDLINE | ID: mdl-39310105

ABSTRACT

Background: Glioblastoma (GBM) is characterized by abundant neovascularization as an essential hallmark. Vasculogenic mimicry (VM) is a predominant pattern of GBM neovascularization. However, the biological functions of circRNAs prompting VM formation in GBM remains unclarified. Methods: The circular RNA circCMTM3 was identified through high-throughput sequencing and bioinformatics analysis. The expression of circCMTM3 in exosomes in glioma tissues and cells was verified via RT-qPCR and FISH. In vitro and in vivo assays, such as EdU, MTS, Transwell, and tube formation assays were performed to investigate functional roles of circCMTM3. Meanwhile, in situ tumorigenesis assay were implemented to explore the influences of circCMTM3 on the GBM progression. Additionally, RNA pull-down, RIP, ChIP, and dual-luciferase reporter gene assays were executed to confirm the underlying regulation mechanism of circCMTM3. Results: CircCMTM3, as a novel circular RNA, was packaged into exosomes derived from glioblastoma stem cells (GSCs), which facilitates the phenotypic transition of differentiated glioma cells (DGCs) to VM. Mechanistically, exosomal circCMTM3 is internalized by DGCs and disrupt the ubiquitination degradation of STAT5A and STAT5B by E3 ubiquitin ligase CNOT4. Additionally, through molecular scaffold function of circCMTM3, STAT5A is activated and triggers transcriptional regulation of target genes including the pro-vasculogenic factor CHI3L2 and the RNA-binding protein SRSF1. Subsequently, circCMTM3/STAT5A/SRSF1 positive feedback loop sustainably enhances VM formation and accelerates tumor progression in GBM. Conclusion: Exosomal circCMTM3 possessing growth factor-mimetic property activates the JAK2/STAT5A pathway via non-canonical manner, and promotes VM formation in GBM. The molecular communications between GSCs and DGCs offers a therapeutic strategy for targeting the neovascularization of GBM.


Subject(s)
Brain Neoplasms , Exosomes , Glioblastoma , Neoplastic Stem Cells , Neovascularization, Pathologic , RNA, Circular , STAT5 Transcription Factor , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , RNA, Circular/metabolism , RNA, Circular/genetics , Humans , STAT5 Transcription Factor/metabolism , Exosomes/metabolism , Neovascularization, Pathologic/metabolism , Cell Line, Tumor , Animals , Neoplastic Stem Cells/metabolism , Mice , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Phosphorylation , Mice, Nude , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Tumor Suppressor Proteins
14.
Cell Biochem Funct ; 42(7): e4118, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39267363

ABSTRACT

Low back pain significantly impacts individuals' quality of life, with intervertebral disc degeneration (IDD) being a primary contributor to this condition. Currently, IDD treatment primarily focuses on symptom management and does not achieve a definitive cure. The cartilage endplate (CEP), a crucial nutrient-supplying tissue of the intervertebral disc, plays a pivotal role in disc degeneration. This review examines the mechanisms underlying CEP degeneration, summarizing recent advancements in understanding the structure and function of CEP, the involvement of various signaling pathways, and the roles of cartilage endplate stem cells (CESCs) and exosomes (Exos) in this process. The aim of this review is to provide a comprehensive reference for future research on CEP. Despite progress in understanding the role of CEP in IDD, the mechanisms underlying CEP degeneration remain incompletely elucidated. Future research poses significant challenges, necessitating further investigations to elucidate the complexities of CEP.


Subject(s)
Cartilage , Intervertebral Disc Degeneration , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/metabolism , Humans , Cartilage/metabolism , Cartilage/pathology , Animals , Intervertebral Disc/pathology , Intervertebral Disc/metabolism , Exosomes/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Stem Cells/pathology , Signal Transduction
15.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273098

ABSTRACT

Osteoarthritis (OA) is a leading cause of pain and disability worldwide in elderly people. There is a critical need to develop novel therapeutic strategies that can effectively manage pain and disability to improve the quality of life for older people. Mesenchymal stem cells (MSCs) have emerged as a promising cell-based therapy for age-related disorders due to their multilineage differentiation and strong paracrine effects. Notably, MSC-derived exosomes (MSC-Exos) have gained significant attention because they can recapitulate MSCs into therapeutic benefits without causing any associated risks compared with direct cell transplantation. These exosomes help in the transport of bioactive molecules such as proteins, lipids, and nucleic acids, which can influence various cellular processes related to tissue repair, regeneration, and immune regulation. In this review, we have provided an overview of MSC-Exos as a considerable treatment option for osteoarthritis. This review will go over the underlying mechanisms by which MSC-Exos may alleviate the pathological hallmarks of OA, such as cartilage degradation, synovial inflammation, and subchondral bone changes. Furthermore, we have summarized the current preclinical evidence and highlighted promising results from in vitro and in vivo studies, as well as progress in clinical trials using MSC-Exos to treat OA.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Osteoarthritis , Exosomes/metabolism , Exosomes/transplantation , Humans , Osteoarthritis/therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Mesenchymal Stem Cells/metabolism , Animals , Mesenchymal Stem Cell Transplantation/methods
16.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273428

ABSTRACT

Diabetic cardiomyopathy (DCM) represents one of the typical complications associated with diabetes. It has been described as anomalies in heart function and structure, with consequent high morbidity and mortality. DCM development can be described by two stages; the first is characterized by left ventricular hypertrophy and diastolic dysfunction, and the second by heart failure (HF) with systolic dysfunction. The proposed mechanisms involve cardiac inflammation, advanced glycation end products (AGEs) and angiotensin II. Furthermore, different studies have focused their attention on cardiomyocyte death through the different mechanisms of programmed cell death, such as apoptosis, autophagy, necrosis, pyroptosis and ferroptosis. Exosome release, adipose epicardial tissue and aquaporins affect DCM development. This review will focus on the description of the mechanisms involved in DCM progression and development.


Subject(s)
Adipose Tissue , Diabetic Cardiomyopathies , Exosomes , Fibrosis , Pericardium , Humans , Exosomes/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Pericardium/metabolism , Pericardium/pathology , Cell Death , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Epicardial Adipose Tissue
17.
Cardiovasc Diabetol ; 23(1): 331, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252021

ABSTRACT

BACKGROUND: Visceral adipose tissue in individuals with obesity is an independent cardiovascular risk indicator. However, it remains unclear whether adipose tissue influences common cardiovascular diseases, such as atherosclerosis, through its secreted exosomes. METHODS: The exosomes secreted by adipose tissue from diet-induced obesity mice were isolated to examine their impact on the progression of atherosclerosis and the associated mechanism. Endothelial apoptosis and the proliferation and migration of vascular smooth muscle cells (VSMCs) within the atherosclerotic plaque were evaluated. Statistical significance was analyzed using GraphPad Prism 9.0 with appropriate statistical tests. RESULTS: We demonstrate that adipose tissue-derived exosomes (AT-EX) exacerbate atherosclerosis progression by promoting endothelial apoptosis, proliferation, and migration of VSMCs within the plaque in vivo. MicroRNA-132/212 (miR-132/212) was detected within AT-EX cargo. Mechanistically, miR-132/212-enriched AT-EX exacerbates palmitate acid-induced endothelial apoptosis via targeting G protein subunit alpha 12 and enhances platelet-derived growth factor type BB-induced VSMC proliferation and migration by targeting phosphatase and tensin homolog in vitro. Importantly, melatonin decreases exosomal miR-132/212 levels, thereby mitigating the pro-atherosclerotic impact of AT-EX. CONCLUSION: These data uncover the pathological mechanism by which adipose tissue-derived exosomes regulate the progression of atherosclerosis and identify miR-132/212 as potential diagnostic and therapeutic targets for atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Exosomes , Mice, Inbred C57BL , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Cell Proliferation/drug effects , Apoptosis/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Cell Movement/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Male , Signal Transduction , Cells, Cultured , Obesity/metabolism , Obesity/pathology , Mice, Knockout, ApoE , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/drug effects , Aortic Diseases/pathology , Aortic Diseases/metabolism , Aortic Diseases/genetics , Becaplermin/pharmacology , Becaplermin/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Mice , Humans
18.
J Orthop Surg Res ; 19(1): 554, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252098

ABSTRACT

BACKGROUND: Facilitating the healing process of injured anterior cruciate ligament (ACL) tissue is crucial for patients to safely return to sports. Stem cell derived exosomes have shown positive effects on enhancing the regeneration of injured tendons/ligaments. However, clinical application of exosomes in terms of storage and pre-assembly is challenging. We hypothesized that lyophilized exosomes derived from human umbilical cord stem cells (hUSC-EX) could enhance the cell activity of chronically injured ACL cells. MATERIALS AND METHODS: We harvested the 8 weeks injured ACL cells from rabbit under IACUC (No. 110232) approval. The studied exosomes were purified from the culture medium of human umbilical cord stem cells (IRB approval No. A202205014), lyophilized to store, and hydrated for use. We compared exosome treated cells with non-exosome treated cells (control group) from the same rabbits. We examined the cell viability, proliferation, migration capability and gene expression of type I and III collagen, TGFß, VEGF, and tenogenesis in the 8 weeks injured ACL cells after hUSC-EX treatment. RESULTS: After hydration, the average size of hUSC-EX was 84.5 ± 70.6 nm, and the cells tested positive for the Alix, TSG101, CD9, CD63, and CD81 proteins but negative for the α-Tubulin protein. After 24 h of treatment, hUSC-EX significantly improved the cell viability, proliferation and migration capability of 8 weeks injured ACL cells compared to that of no exosome treatment group. In addition, the expression of collagen synthesis, TGFß, VEGF, and tenogenesis gene were all significantly increased in the 8 weeks injured ACL cells after 24 h hUSC-EX delivery. DISCUSSION: Lyophilized exosomes are easily stored and readily usable after hydration, thereby preserving their characteristic properties. Treatment with lyophilized hUSC-EX improved the activity and gene expression of 8 weeks injured ACL cells. CONCLUSION: Lyophilized hUSC-EX preserve the characteristics of exosomes and can improve chronically injured (8 weeks) ACL cells. Lyophilized hUSC-EX could serve as effective and safe biomaterials that are ready to use at room temperature to enhance cell activity in patients with partial ACL tears and after remnant preservation ACL reconstruction.


Subject(s)
Anterior Cruciate Ligament Injuries , Exosomes , Animals , Rabbits , Exosomes/metabolism , Anterior Cruciate Ligament Injuries/therapy , Humans , Freeze Drying , Cell Proliferation , Umbilical Cord/cytology , Cells, Cultured , Cell Survival/physiology , Cell Movement/physiology , Chronic Disease
19.
Int J Nanomedicine ; 19: 9035-9053, 2024.
Article in English | MEDLINE | ID: mdl-39253060

ABSTRACT

Background: Ischemic preconditioning-induced serum exosomes (IPC-exo) protected rat heart against myocardial ischemia/reperfusion injury. However, whether IPC-exo regulate replacement fibrosis after myocardial infarction (MI) and the underlying mechanisms remain unclear. MicroRNAs (miRs) are important cargos of exosomes and play an essential role in cardioprotection. We aim to investigate whether IPC-exo regulate post-MI replacement fibrosis by transferring cardioprotective miRs and its action mechanism. Methods: Exosomes obtained from serum of adult rats in control (Con-exo) and IPC groups were identified and analyzed, subsequently intracardially injected into MI rats following ligation. Their miRs profiles were identified using high-throughput miR sequencing to identify target miRs for bioinformatics analysis. Luciferase reporter assays confirmed target genes of selected miRs. IPC-exo transfected with selected miRs antagomir or NC were intracardially administered to MI rats post-ligation. Cardiac function and degree of replacement fibrosis were detected 4 weeks post-MI. Results: IPC-exo exerted cardioprotective effects against excessive replacement fibrosis. MiR sequencing and RT-qPCR identified miR-133a-3p as most significantly different between IPC-exo and Con-exo. MiR-133a-3p directly targeted latent transforming growth factor beta binding protein 1 (LTBP1) and protein phosphatase 2, catalytic subunit, alpha isozyme (PPP2CA). KEGG analysis showed that transforming growth factor-ß (TGF-ß) was one of the most enriched signaling pathways with miR-133a-3p. Comparing to injection of IPC-exo transfected with miR-133a-3p antagomir NC, injecting IPC-exo transfected with miR-133a-3p antagomir abolished protective effects of IPC-exo on declining excessive replacement fibrosis and cardiac function enhancement, while increasing the messenger RNA and protein expression of LTBP1, PPP2CA, and TGF-ß1in MI rats. Conclusion: IPC-exo inhibit excessive replacement fibrosis and improve cardiac function post-MI by transferring miR-133a-3p, the mechanism is associated with directly targeting LTBP1 and PPP2CA, and indirectly regulating TGF-ß pathway in rats. Our finding provides potential therapeutic effect of IPC-induced exosomal miR-133a-3p for cardiac repair.


Subject(s)
Exosomes , MicroRNAs , Myocardial Infarction , Protein Phosphatase 2 , Animals , MicroRNAs/blood , MicroRNAs/genetics , Myocardial Infarction/blood , Myocardial Infarction/therapy , Myocardial Infarction/genetics , Exosomes/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Male , Rats , Rats, Sprague-Dawley , Fibrosis , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/therapy , Myocardium/metabolism , Ischemic Preconditioning/methods , Ischemic Preconditioning, Myocardial/methods
20.
Eur J Pharm Biopharm ; 203: 114460, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39218361

ABSTRACT

Glioblastoma (GBM) stands for the most common and aggressive type of brain tumour in adults. It is highly invasive, which explains its short rate of survival. Little is known about its risk factors, and current therapy is still ineffective. Hence, efforts are underway to develop novel and effective treatment approaches against this type of cancer. Exosomes are being explored as a promising strategy for conveying and delivering therapeutic cargo to GBM cells. They can fuse with the GBM cell membrane and, consequently, serve as delivery systems in this context. Due to their nanoscale size, exosomes can cross the blood-brain barrier (BBB), which constitutes a significant hurdle to most chemotherapeutic drugs used against GBM. They can subsequently inhibit oncogenes, activate tumour suppressor genes, induce immune responses, and control cell growth. However, despite representing a promising tool for the treatment of GBM, further research and clinical studies regarding exosome biology, engineering, and clinical applications still need to be completed. Here, we sought to review the application of exosomes in the treatment of GBM through an in-depth analysis of the scientific and clinical studies on the entire process, from the isolation and purification of exosomes to their design and transformation into anti-oncogenic drug delivery systems. Surface modification of exosomes to enhance BBB penetration and GBM-cell targeting is also a topic of discussion.


Subject(s)
Antineoplastic Agents , Blood-Brain Barrier , Brain Neoplasms , Drug Delivery Systems , Exosomes , Glioblastoma , Exosomes/metabolism , Glioblastoma/drug therapy , Glioblastoma/therapy , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Drug Delivery Systems/methods , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL