Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.160
Filter
1.
Cell Commun Signal ; 22(1): 344, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937789

ABSTRACT

BACKGROUND: Tumor cells release extracellular vesicles (EVs) that contribute to the polarization of macrophages towards tumor-associated macrophages (TAMs). High expression levels of the RNA binding protein IGF2BP2/IMP2 are correlated with increased tumor cell proliferation, invasion, and poor prognosis in the clinic. However, there is a lack of understanding of whether IMP2 affects the cargo of cancer cell-derived EVs, thereby modulating macrophage polarization. METHODS: EVs were isolated from IMP2-expressing HCT116 parental cells (WT) and CRISPR/Cas9 IMP2 knockout (KO) cells. EVs were characterized according to MISEV guidelines, microRNA cargo was assessed by microRNA-Seq, and the protein cargo was analyzed by proteomics. Primary human monocyte-derived macrophages (HMDMs) were polarized by EVs, and the expression of genes and surface markers was assessed using qPCR and flow cytometry, respectively. Morphological changes of macrophages, as well as the migratory potential of cancer cells, were assessed by the Incucyte® system and macrophage matrix degradation potential by zymography. Changes in the metabolic activity of macrophages were quantified using a Seahorse® analyzer. For in vivo studies, EVs were injected into the yolk sac of zebrafish larvae, and macrophages were isolated by fluorescence-activated cell sorting. RESULTS: EVs from WT and KO cells had a similar size and concentration and were positive for 25 vesicle markers. The expression of tumor-promoting genes was higher in macrophages polarized with WT EVs than KO EVs, while the expression of TNF and IL6 was reduced. A similar pattern was observed in macrophages from zebrafish larvae treated in vivo. WT EV-polarized macrophages showed a higher abundance of TAM-like surface markers, higher matrix degrading activity, as well as a higher promotion of cancer cell migration. MicroRNA-Seq revealed a significant difference in the microRNA composition of WT and KO EVs, particularly a high abundance of miR-181a-5p in WT EVs, which was absent in KO EVs. Inhibitors of macropinocytosis and phagocytosis antagonized the delivery of miR-181a-5p into macrophages and the downregulation of the miR-181a-5p target DUSP6. Proteomics data showed differences in protein cargo in KO vs. WT EVs, with the differentially abundant proteins mainly involved in metabolic pathways. WT EV-treated macrophages exhibited a higher basal oxygen consumption rate and a lower extracellular acidification rate than KO EV-treated cells. CONCLUSION: Our results show that IMP2 determines the cargo of EVs released by cancer cells, thereby modulating the EVs' actions on macrophages. Expression of IMP2 is linked to the secretion of EVs that polarize macrophages towards a tumor-promoting phenotype.


Subject(s)
Extracellular Vesicles , RNA-Binding Proteins , Tumor-Associated Macrophages , Zebrafish , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Animals , Tumor-Associated Macrophages/metabolism , HCT116 Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Movement/genetics , Macrophages/metabolism
2.
Cells ; 13(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38920665

ABSTRACT

Pleural mesothelioma (PM) is a highly aggressive tumor that is caused by asbestos exposure and lacks effective therapeutic regimens. Current procedures for PM diagnosis are invasive and can take a long time to reach a definitive result. Small extracellular vesicles (sEVs) have been identified as important communicators between tumor cells and their microenvironment via their cargo including circular RNAs (circRNAs). CircRNAs are thermodynamically stable, highly conserved, and have been found to be dysregulated in cancer. This study aimed to identify potential biomarkers for PM diagnosis by investigating the expression of specific circRNA gene pattern (hsa_circ_0007386) in cells and sEVs using digital polymerase chain reaction (dPCR). For this reason, 5 PM, 14 non-PM, and one normal mesothelial cell line were cultured. The sEV was isolated from the cells using the gold standard ultracentrifuge method. The RNA was extracted from both cells and sEVs, cDNA was synthesized, and dPCR was run. Results showed that hsa_circ_0007386 was significantly overexpressed in PM cell lines and sEVs compared to non-PM and normal mesothelial cell lines (p < 0.0001). The upregulation of hsa_circ_0007386 in PM highlights its potential as a diagnostic biomarker. This study underscores the importance and potential of circRNAs and sEVs as cancer diagnostic tools.


Subject(s)
Biomarkers, Tumor , Extracellular Vesicles , Mesothelioma , RNA, Circular , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Mesothelioma/genetics , Mesothelioma/diagnosis , Cell Line, Tumor , Pleural Neoplasms/genetics , Pleural Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , Mesothelioma, Malignant/genetics , Mesothelioma, Malignant/diagnosis
3.
Signal Transduct Target Ther ; 9(1): 151, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38910148

ABSTRACT

Pancreatic cancer is one of the deadly malignancies with a significant mortality rate and there are currently few therapeutic options for it. The tumor microenvironment (TME) in pancreatic cancer, distinguished by fibrosis and the existence of cancer-associated fibroblasts (CAFs), exerts a pivotal influence on both tumor advancement and resistance to therapy. Recent advancements in the field of engineered extracellular vesicles (EVs) offer novel avenues for targeted therapy in pancreatic cancer. This study aimed to develop engineered EVs for the targeted reprogramming of CAFs and modulating the TME in pancreatic cancer. EVs obtained from bone marrow mesenchymal stem cells (BMSCs) were loaded with miR-138-5p and the anti-fibrotic agent pirfenidone (PFD) and subjected to surface modification with integrin α5-targeting peptides (named IEVs-PFD/138) to reprogram CAFs and suppress their pro-tumorigenic effects. Integrin α5-targeting peptide modification enhanced the CAF-targeting ability of EVs. miR-138-5p directly inhibited the formation of the FERMT2-TGFBR1 complex, inhibiting TGF-ß signaling pathway activation. In addition, miR-138-5p inhibited proline-mediated collagen synthesis by directly targeting the FERMT2-PYCR1 complex. The combination of miR-138-5p and PFD in EVs synergistically promoted CAF reprogramming and suppressed the pro-cancer effects of CAFs. Preclinical experiments using the orthotopic stroma-rich and patient-derived xenograft mouse models yielded promising results. In particular, IEVs-PFD/138 effectively reprogrammed CAFs and remodeled TME, which resulted in decreased tumor pressure, enhanced gemcitabine perfusion, tumor hypoxia amelioration, and greater sensitivity of cancer cells to chemotherapy. Thus, the strategy developed in this study can improve chemotherapy outcomes. Utilizing IEVs-PFD/138 as a targeted therapeutic agent to modulate CAFs and the TME represents a promising therapeutic approach for pancreatic cancer.


Subject(s)
Cancer-Associated Fibroblasts , Extracellular Vesicles , MicroRNAs , Pancreatic Neoplasms , Tumor Microenvironment , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Mice , MicroRNAs/genetics , Animals , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Cellular Reprogramming/genetics , Cellular Reprogramming/drug effects , Cell Line, Tumor , Mesenchymal Stem Cells/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Gemcitabine
4.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928163

ABSTRACT

Obesity is a risk factor for type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). Adipose tissue (AT) extracellular vesicles (EVs) could play a role in obesity and T2DM associated CVD progression via the influence of their specific cargo on gene expression in recipient cells. The aim of this work was to evaluate the effects of AT EVs of patients with obesity with/without T2DM on reverse cholesterol transport (RCT)-related gene expression in human monocyte-derived macrophages (MDMs) from healthy donors. AT EVs were obtained after ex vivo cultivation of visceral and subcutaneous AT (VAT and SAT, respectively). ABCA1, ABCG1, PPARG, LXRß (NR1H2), and LXRα (NR1H3) mRNA levels in MDMs as well as in origine AT were determined by a real-time PCR. T2DM VAT and SAT EVs induced ABCG1 gene expression whereas LXRα and PPARG mRNA levels were simultaneously downregulated. PPARG mRNA levels also decreased in the presence of VAT EVs of obese patients without T2DM. In contrast ABCA1 and LXRß mRNA levels tended to increase with the addition of obese AT EVs. Thus, AT EVs can influence RCT gene expression in MDMs during obesity, and the effects are dependent on T2DM status.


Subject(s)
ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Adipose Tissue , Cholesterol , Diabetes Mellitus, Type 2 , Extracellular Vesicles , Liver X Receptors , Macrophages , Obesity , PPAR gamma , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Obesity/metabolism , Obesity/genetics , Liver X Receptors/metabolism , Liver X Receptors/genetics , Macrophages/metabolism , Cholesterol/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Adipose Tissue/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Female , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Male , Middle Aged , Biological Transport , Gene Expression Regulation , Adult , RNA, Messenger/metabolism , RNA, Messenger/genetics
5.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928399

ABSTRACT

The ability of tumor-derived extracellular vesicles (EVs) to modulate the function of myeloid cells is widely recognized. Hence, a comprehensive understanding of the distinct components associated with EVs and the signals that they deliver to myeloid cells could provide potential approaches to impede the immunosuppression by myeloid-derived suppressor cells (MDSCs). We investigated melanoma EV-associated microRNAs (miRs) using the RET transgenic melanoma mouse model and simulated their transfer to normal myeloid cells by transfecting immature mouse myeloid cells and human monocytes. We observed elevated levels of miR-125a-5p, -125b-5p, and let-7e-5p in mouse melanoma-infiltrating MDSCs. In addition, miR-125a-5p levels in the tumor microenvironment correlated with mouse melanoma progression. The delivery of miR-125a-5p, alone or in combination with let-7e-5p and miR-99b-5p from the same genomic cluster, to normal myeloid cells resulted in their conversion to MDSC-like cells. Our findings indicate that miR-125a-5p could modulate myeloid cell activation in the melanoma microenvironment via a NF-κB-dependent mechanism.


Subject(s)
Melanoma , MicroRNAs , Myeloid-Derived Suppressor Cells , Tumor Microenvironment , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Animals , Humans , Mice , Tumor Microenvironment/genetics , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice, Transgenic , NF-kappa B/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mice, Inbred C57BL , Monocytes/metabolism
6.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928481

ABSTRACT

Ischemic stroke is a major cause of mortality worldwide. Proper etiological subtyping of ischemic stroke is crucial for tailoring treatment strategies. This study explored the utility of circulating microRNAs encapsulated in extracellular vesicles (EV-miRNAs) to distinguish the following ischemic stroke subtypes: large artery atherosclerosis (LAA), cardioembolic stroke (CES), and small artery occlusion (SAO). Using next-generation sequencing (NGS) and machine-learning techniques, we identified differentially expressed miRNAs (DEMs) associated with each subtype. Through patient selection and diagnostic evaluation, a cohort of 70 patients with acute ischemic stroke was classified: 24 in the LAA group, 24 in the SAO group, and 22 in the CES group. Our findings revealed distinct EV-miRNA profiles among the groups, suggesting their potential as diagnostic markers. Machine-learning models, particularly logistic regression models, exhibited a high diagnostic accuracy of 92% for subtype discrimination. The collective influence of multiple miRNAs was more crucial than that of individual miRNAs. Additionally, bioinformatics analyses have elucidated the functional implications of DEMs in stroke pathophysiology, offering insights into the underlying mechanisms. Despite limitations like sample size constraints and retrospective design, our study underscores the promise of EV-miRNAs coupled with machine learning for ischemic stroke subtype classification. Further investigations are warranted to validate the clinical utility of the identified EV-miRNA biomarkers in stroke patients.


Subject(s)
Biomarkers , Circulating MicroRNA , Exosomes , Ischemic Stroke , Machine Learning , Humans , Ischemic Stroke/blood , Ischemic Stroke/genetics , Ischemic Stroke/diagnosis , Male , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Female , Aged , Middle Aged , Exosomes/genetics , Exosomes/metabolism , Biomarkers/blood , High-Throughput Nucleotide Sequencing/methods , Computational Biology/methods , MicroRNAs/blood , MicroRNAs/genetics , Gene Expression Profiling/methods , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics
7.
Anticancer Res ; 44(7): 2981-2988, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925824

ABSTRACT

BACKGROUND/AIM: Extracellular vesicle DNA (EV-DNA) has emerged as a novel biomarker for tumor mutation detection using liquid biopsies, exhibiting biological advantages compared to cell-free DNA (cfDNA). This study assessed the feasibility of EV-DNA and cfDNA extraction and sequencing in old serum samples of patients with breast cancer (BC). PATIENTS AND METHODS: A total of 28 serum samples of 27 patients with corresponding clinical information were collected between 1983 and 1991. EV-DNA was extracted using Exo-GAG kit (Nasabiotech) and cfDNA using QIAsymphony DSP Virus/Pathogen Midi Kit (Qiagen), respectively. Subsequently, 10 matched samples (EV-DNA n=5, cfDNA n=5) of five patients were subjected to sequencing using the Oncomine™ Breast cfDNA Research Assay v2 (Thermo Fisher Scientific). RESULTS: Samples were collected on median 1.9 years after primary diagnosis [interquartile range (IQR)=0.2-7.2]. Median follow-up was 9.5 years (IQR=5.2-14.2). Median age of serum samples was 36.1 years (IQR=34.5-37.3). EV-DNA and cfDNA were extracted from 100% (28/28) of the included samples. Both, DNA quantity and concentration were comparable between EV-DNA and cfDNA. Sequencing was successfully performed in 100% (10/10) of the included samples. Two matched analyses yielded equivalent results in EV-DNA and cfDNA (no mutations, n=1; PIK3CA mutation, n=1), whilst in two analyses, PIK3CA mutation was only found in cfDNA, and in one analysis, a TP53 mutation was only found in EV-DNA. CONCLUSION: EV-DNA extraction and sequencing in old serum samples of patients with BC is feasible and has the potential to address clinically relevant questions in longitudinal studies.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Extracellular Vesicles , Humans , Breast Neoplasms/genetics , Breast Neoplasms/blood , Female , Extracellular Vesicles/genetics , Adult , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Mutation , Middle Aged , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Liquid Biopsy/methods , Sequence Analysis, DNA/methods
8.
Cancer Biol Ther ; 25(1): 2364433, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38926911

ABSTRACT

Prostate cancer has heterogeneous growth patterns, and its prognosis is the poorest when it progresses to a neuroendocrine phenotype. Using bioinformatic analysis, we evaluated RNA expression of neuroendocrine genes in a panel of five different cancer types: prostate adenocarcinoma, breast cancer, kidney chromophobe, kidney renal clear cell carcinoma and kidney renal papillary cell carcinoma. Our results show that specific neuroendocrine genes are significantly dysregulated in these tumors, suggesting that they play an active role in cancer progression. Among others, synaptophysin (SYP), a conventional neuroendocrine marker, is upregulated in prostate adenocarcinoma (PRAD) and breast cancer (BRCA). Our analysis shows that SYP is enriched in small extracellular vesicles (sEVs) derived from plasma of PRAD patients, but it is absent in sEVs derived from plasma of healthy donors. Similarly, classical sEV markers are enriched in sEVs derived from plasma of prostate cancer patients, but weakly detectable in sEVs derived from plasma of healthy donors. Overall, our results pave the way to explore new strategies to diagnose these diseases based on the neuroendocrine gene expression in patient tumors or plasma sEVs.


Subject(s)
Adenocarcinoma , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Synaptophysin/metabolism , Synaptophysin/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Gene Expression Profiling/methods
9.
Genes (Basel) ; 15(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38927671

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease targeting the brain and spinal cord. Non-neuronal cells, including macrophages, may contribute to the disruption of motor neurons (MNs), neuromuscular junction dismantling and clinical signs of ALS. Understanding the modality and the effect of MNs-macrophage communication is pivotal. Here, we focus on extracellular vesicle (EVS)-mediated communication and, in particular, we analyze the response of macrophages. NSC-34 cells transfected with mutant SOD1 (G93A, A4V, G85R, G37R) and differentiated towards MN-like cells, and Raw 264.7 macrophages are the cellular models of the study. mSOD1 NSC-34 cells release a high number of vesicles, both large-lEVs (300 nm diameter) and small-sEVs (90 nm diameter), containing inflammation-modulating molecules, and are efficiently taken up by macrophages. RT-PCR analysis of inflammation mediators demonstrated that the conditioned medium of mSOD1 NSC-34 cells polarizes Raw 264.7 macrophages towards both pro-inflammatory and anti-inflammatory phenotypes. sEVs act on macrophages in a time-dependent manner: an anti-inflammatory response mediated by TGFß firstly starts (12 h); successively, the response shifts towards a pro-inflammation IL-1ß-mediated (48 h). The response of macrophages is strictly dependent on the SOD1 mutation type. The results suggest that EVs impact physiological and behavioral macrophage processes and are of potential relevance to MN degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis , Extracellular Vesicles , Macrophages , Motor Neurons , Superoxide Dismutase-1 , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Mice , RAW 264.7 Cells , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Macrophages/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mutation , Transfection , Humans
10.
Med Oncol ; 41(7): 169, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839666

ABSTRACT

To investigate extracellular vesicles (EVs), biomarkers for predicting lymph node invasion (LNI) in patients with high-risk prostate cancer (HRPCa), plasma, and/or urine samples were prospectively collected from 45 patients with prostate cancer (PCa) and five with benign prostatic hyperplasia (BPH). Small RNA sequencing was performed to identify miRNAs in the EVs. All patients with PCa underwent radical prostatectomy and extended pelvic lymph node dissection. Differentially expressed miRNAs were identified in patients with and without pathologically-verified LNI. The candidate miRNAs were validated in low-risk prostate cancer (LRPCa) and BPH. Four miRNA species (e.g., miR-126-3p) and three miRNA species (e.g., miR-27a-3p) were more abundant in urinary and plasma EVs, respectively, of patients with PCa. None of these miRNA species were shared between urinary and plasma EVs. miR-126-3p was significantly more abundant in patients with HR PCa with LNI than in those without (P = 0.018). miR-126-3p was significantly more abundant in the urinary EVs of patients with HRPCa than in those with LRPCa (P = 0.017) and BPH (P = 0.011). In conclusion, urinary EVs-derived miR-126-3p may serve as a good biomarker for predicting LNI in patients with HRPCa.


Subject(s)
Biomarkers, Tumor , Extracellular Vesicles , Lymphatic Metastasis , MicroRNAs , Prostatic Neoplasms , Humans , Male , MicroRNAs/urine , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/urine , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Aged , Middle Aged , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/urine , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/urine , Lymph Nodes/pathology , Prostatectomy , Prospective Studies
11.
Biomed Pharmacother ; 176: 116868, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850647

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic relapsing disease of the gastrointestinal (GI) system that includes two groups, Crohn's disease (CD) and ulcerative colitis (UC). To cope with these two classes of IBD, the investigation of pathogenic mechanisms and the discovery of new diagnostic and therapeutic approaches are crucial. Long non-coding RNAs (lncRNAs) which are non-coding RNAs with a length of longer than 200 nucleotides have indicated significant association with the pathology of IBD and strong potential to be used as accurate biomarkers in diagnosing and predicting responses to the IBD treatment. In the current review, we aim to investigate the role of lncRNAs in the pathology and development of IBD. We first describe recent advances in research on dysregulated lncRNAs in the pathogenesis of IBD from the perspective of epithelial barrier function, intestinal immunity, mitochondrial function, and intestinal autophagy. Then, we highlight the possible translational role of lncRNAs as therapeutic targets, diagnostic biomarkers, and predictors of therapeutic response in colon tissues and plasma samples. Finally, we discuss the potential of extracellular vesicles and their lncRNA cargo in the pathophysiology, diagnosis, and treatment of IBD.


Subject(s)
Biomarkers , Extracellular Vesicles , Inflammatory Bowel Diseases , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Biomarkers/metabolism , Biomarkers/blood , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/therapy , Animals , Colitis, Ulcerative/genetics , Colitis, Ulcerative/diagnosis
12.
Sci Rep ; 14(1): 13560, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866905

ABSTRACT

L1CAM-positive extracellular vesicles (L1EV) are an emerging biomarker that may better reflect ongoing neuronal damage than other blood-based biomarkers. The physiological roles and regulation of L1EVs and their small RNA cargoes following stroke is unknown. We sought to characterize L1EV small RNAs following stroke and assess L1EV RNA signatures for diagnosing stroke using weighted gene co-expression network analysis and random forest (RF) machine learning algorithms. Interestingly, small RNA sequencing of plasma L1EVs from patients with stroke and control patients (n = 28) identified micro(mi)RNAs known to be enriched in the brain. Weighted gene co-expression network analysis (WGCNA) revealed small RNA transcript modules correlated to diagnosis, initial NIH stroke scale, and age. L1EV RNA signatures associated with the diagnosis of AIS were derived from WGCNA and RF classification. These small RNA signatures demonstrated a high degree of accuracy in the diagnosis of AIS with an area under the curve (AUC) of the signatures ranging from 0.833 to 0.932. Further work is necessary to understand the role of small RNA L1EV cargoes in the response to brain injury, however, this study supports the utility of L1EV small RNA signatures as a biomarker of stroke.


Subject(s)
Biomarkers , Extracellular Vesicles , Ischemic Stroke , Neural Cell Adhesion Molecule L1 , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Male , Ischemic Stroke/genetics , Ischemic Stroke/metabolism , Ischemic Stroke/blood , Ischemic Stroke/diagnosis , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Female , Aged , Biomarkers/blood , Middle Aged , Machine Learning , MicroRNAs/genetics , MicroRNAs/blood , MicroRNAs/metabolism
13.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891921

ABSTRACT

The involvement of the microRNA miR165a in the light-dependent mechanisms of regulation of target genes in maize (Zea mays) has been studied. The light-induced change in the content of free miR165a was associated with its binding by the AGO10 protein and not with a change in the rate of its synthesis from the precursor. The use of knockout Arabidopsis plants for the phytochrome A and B genes demonstrated that the presence of an active form of phytochrome B causes an increase in the level of the RNA-induced silencing miR165a complex, which triggers the degradation of target mRNAs. The two fractions of vesicles from maize leaves, P40 and P100 that bind miR165a, were isolated by ultracentrifugation. The P40 fraction consisted of larger vesicles of the size >0.170 µm, while the P100 fraction vesicles were <0.147 µm. Based on the quantitative PCR data, the predominant location of miR165a on the surface of extracellular vesicles of both fractions was established. The formation of the active form of phytochrome upon the irradiation of maize plants with red light led to a redistribution of miR165a, resulting in an increase in its proportion inside P40 vesicles and a decrease in P100 vesicles.


Subject(s)
Light , MicroRNAs , Phytochrome , Plant Leaves , Signal Transduction , Zea mays , Zea mays/genetics , Zea mays/metabolism , Zea mays/radiation effects , MicroRNAs/genetics , MicroRNAs/metabolism , Plant Leaves/metabolism , Plant Leaves/genetics , Plant Leaves/radiation effects , Phytochrome/metabolism , Phytochrome/genetics , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/radiation effects , Phytochrome A/metabolism , Phytochrome A/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Phytochrome B/metabolism , Phytochrome B/genetics
15.
Curr Aging Sci ; 17(2): 118-126, 2024.
Article in English | MEDLINE | ID: mdl-38904154

ABSTRACT

Containing information molecules from their parent cells and inclining to fuse with targeted cells, bone marrow mesenchymal stromal cells-derived extracellular vesicles (MSCs- EV) are valuable in nanomedicine. BACKGROUND: The effects of aging on the paracrine mechanism and in the production and action of MSCs-EV and their cargos of miR-26a and siRNA-26a for the treatment of tubular renal cells under nephrotoxicity injury remain unelucidated. OBJECTIVE: The purpose of this study was to evaluate MSCs-EV of different ages and their ability to deliver the cargos of miR-26a and siRNA-26a to target renal tubular cells affected by nephrotoxicity injury. METHODS: In a model of gentamicin-induced nephrotoxicity, renal tubular cells treated with MSCs-EV expressing or not expressing microRNA-26a were analyzed. Western blotting was utilized to evaluate cell cycle markers, and MTT assay was utilized to evaluate auto-renovation capacity. RESULTS: Tubular cells under nephrotoxicity injury showed decreased proliferative capacity, but the treatment in the tubular renal cells under nephrotoxicity injury with MSCs-EV expressing microRNA-26a showed nephroprotective effects, regardless of EV age. While the treatment with EV-mediated siRNA-26a failed to preserve the nephroprotective effects equally, regardless of age. CONCLUSION: Mesenchymal stromal cell nanovesicles carry microRNA with nephroprotective proprieties regardless of aging.


Subject(s)
Cell Proliferation , Kidney Tubules , Mesenchymal Stem Cells , MicroRNAs , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Kidney Tubules/pathology , Kidney Tubules/metabolism , Aging/metabolism , Aging/pathology , Aging/genetics , Gentamicins/toxicity , Gentamicins/adverse effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Age Factors , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Cell Line , Cells, Cultured , Paracrine Communication , Disease Models, Animal , Humans
16.
Methods Mol Biol ; 2822: 101-123, 2024.
Article in English | MEDLINE | ID: mdl-38907915

ABSTRACT

Extracellular vesicles (EV) are rich in small RNA; however, a frequent caveat can be low abundance of EV RNA content, especially in clinical studies. NanoString MicroRNA Assays allow for multiplexed profiling of n = 800 mature microRNAs and can be applied to assess EV microRNA cargo. Here, we describe a method to adapt NanoString nCounter microRNA profiling to assess mature microRNA expression in low-concentration RNA samples, including concentrating the RNA, quantifying the RNA, and performing the NanoString protocol. Twelve samples can be assessed at one time using this method.


Subject(s)
Extracellular Vesicles , MicroRNAs , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , MicroRNAs/genetics , MicroRNAs/analysis , Humans , Gene Expression Profiling/methods
17.
BMC Bioinformatics ; 25(1): 221, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902629

ABSTRACT

BACKGROUND: Extracellular vesicle-derived (EV)-miRNAs have potential to serve as biomarkers for the diagnosis of various diseases. miRNA microarrays are widely used to quantify circulating EV-miRNA levels, and the preprocessing of miRNA microarray data is critical for analytical accuracy and reliability. Thus, although microarray data have been used in various studies, the effects of preprocessing have not been studied for Toray's 3D-Gene chip, a widely used measurement method. We aimed to evaluate batch effect, missing value imputation accuracy, and the influence of preprocessing on measured values in 18 different preprocessing pipelines for EV-miRNA microarray data from two cohorts with amyotrophic lateral sclerosis using 3D-Gene technology. RESULTS: Eighteen different pipelines with different types and orders of missing value completion and normalization were used to preprocess the 3D-Gene microarray EV-miRNA data. Notable results were suppressed in the batch effects in all pipelines using the batch effect correction method ComBat. Furthermore, pipelines utilizing missForest for missing value imputation showed high agreement with measured values. In contrast, imputation using constant values for missing data exhibited low agreement. CONCLUSIONS: This study highlights the importance of selecting the appropriate preprocessing strategy for EV-miRNA microarray data when using 3D-Gene technology. These findings emphasize the importance of validating preprocessing approaches, particularly in the context of batch effect correction and missing value imputation, for reliably analyzing data in biomarker discovery and disease research.


Subject(s)
Extracellular Vesicles , MicroRNAs , Oligonucleotide Array Sequence Analysis , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Oligonucleotide Array Sequence Analysis/methods , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Gene Expression Profiling/methods
18.
Int J Nanomedicine ; 19: 6085-6098, 2024.
Article in English | MEDLINE | ID: mdl-38911502

ABSTRACT

Introduction: Endometriosis (EM) is an estrogen-dependent benign gynecologic disease affecting approximately 10% of reproductive-age women with a high recurrence rate, but lacks reliable biomarkers. No previous studies have investigated the possible use of extracellular vesicle (EV)-associated micro RNAs (miRNAs) from menstrual blood (MB) as candidate diagnostic or prognostic markers of EM. Methods: Specimens were obtained from endometriosis and non-endometriosis patients at the International Peace Maternity and Child Health Hospital in Shanghai. Microarray was used to screen differentially expressed miRNAs among peritoneal fluid (PF), fallopian tube fluid (FF), and MB. Dual-luciferase reporter gene assay was carried out to verify the relationship between miR-4443 and ACSS2. Cell proliferation and Transwell invasion assays were performed in vitro after intervention on miR-4443 and ACSS2 in hEM15A human endometrial stromal cells and primary human endometrial stromal cells (hESCs). Spearman correlation analysis, receiver operating characteristic (ROC) curve analysis, and survival analysis were applied to clinical data, including severity of symptoms and relapse of EM among EM patients. Results: EV-associated miR-4443 was abundant in MB of endometriosis patients. ACSS2 knockdown and miR-4443 overexpression promoted cell proliferation and migration via the PI3K/AKT pathway. miR-4443 levels in MB-EVs were positively correlated with the degree of dyspareunia (r=0.64; P<0.0001) and dysmenorrhea (r=0.42; P<0.01) in the endometriosis group. ROC curve analyses showed an area under the curve (AUC) of 0.741 (95% CI 0.624-0.858; P<0.05) for miR-4443 and an AUC of 0.929 (95% CI 0.880-0.978; P<0.05) for the combination of miR-4443 and dysmenorrhea. Conclusion: MB-derived EV-associated miR-4443 might participate in endometriosis development, thus providing a new candidate biomarker for the noninvasive prediction of endometriosis recurrence.


Subject(s)
Cell Proliferation , Endometriosis , Extracellular Vesicles , MicroRNAs , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Humans , Endometriosis/metabolism , Endometriosis/genetics , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Adult , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Disease Progression , Cell Movement , Signal Transduction , Cell Line , Endometrium/metabolism , Endometrium/pathology
19.
Genomics ; 116(4): 110875, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849018

ABSTRACT

Exploration of a stably expressed gene as a reference is critical for the accurate evaluation of miRNAs isolated from small extracellular vesicles (sEVs). In this study, we analyzed small RNA sequencing on plasma sEV miRNAs in the training dataset (n = 104) and found that miR-140-3p was the most stably expressed candidate reference for sEV miRNAs. We further demonstrated that miR-140-3p expressed most stably in the validation cohort (n = 46) when compared to two other reference miRNAs, miR-451a and miR-1228-3p, and the commonly-used miRNA reference U6. Finally, we compared the capability of miR-140-3p and U6 as the internal reference for sEV miRNA expression by evaluating key miRNAs expression in lung cancer patients and found that miR-140-3p was more suitable as a sEV miRNA reference gene. Taken together, our data indicated miR-140-3p as a stable internal reference miRNA of plasma sEVs to evaluate miRNA expression profiles in lung cancer patients.


Subject(s)
Extracellular Vesicles , Lung Neoplasms , MicroRNAs , Humans , MicroRNAs/blood , MicroRNAs/genetics , Lung Neoplasms/genetics , Lung Neoplasms/blood , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , Male , Reference Standards , Real-Time Polymerase Chain Reaction/standards , Middle Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics
20.
Cell Mol Biol Lett ; 29(1): 82, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822260

ABSTRACT

BACKGROUND: Hepatic stellate cells (HSCs) play a crucial role in the development of fibrosis in non-alcoholic fatty liver disease (NAFLD). Small extracellular vesicles (sEV) act as mediators for intercellular information transfer, delivering various fibrotic factors that impact the function of HSCs in liver fibrosis. In this study, we investigated the role of lipotoxic hepatocyte derived sEV (LTH-sEV) in HSCs activation and its intrinsic mechanisms. METHODS: High-fat diet (HFD) mice model was constructed to confirm the expression of LIMA1. The relationship between LIMA1-enriched LTH-sEV and LX2 activation was evaluated by measurement of fibrotic markers and related genes. Levels of mitophagy were detected using mt-keima lentivirus. The interaction between LIMA1 and PINK1 was discovered through database prediction and molecular docking. Finally, sEV was injected to investigate whether LIMA1 can accelerate HFD induced liver fibrosis in mice. RESULTS: LIMA1 expression was upregulated in lipotoxic hepatocytes and was found to be positively associated with the expression of the HSCs activation marker α-SMA. Lipotoxicity induced by OPA led to an increase in both the level of LIMA1 protein in LTH-sEV and the release of LTH-sEV. When HSCs were treated with LTH-sEV, LIMA1 was observed to hinder LX2 mitophagy while facilitating LX2 activation. Further investigation revealed that LIMA1 derived from LTH-sEV may inhibit PINK1-Parkin-mediated mitophagy, consequently promoting HSCs activation. Knocking down LIMA1 significantly attenuates the inhibitory effects of LTH-sEV on mitophagy and the promotion of HSCs activation. CONCLUSIONS: Lipotoxic hepatocyte-derived LIMA1-enriched sEVs play a crucial role in promoting HSCs activation in NAFLD-related liver fibrosis by negatively regulating PINK1 mediated mitophagy. These findings provide new insights into the pathological mechanisms involved in the development of fibrosis in NAFLD.


Subject(s)
Diet, High-Fat , Extracellular Vesicles , Hepatic Stellate Cells , Hepatocytes , Liver Cirrhosis , Mice, Inbred C57BL , Mitophagy , Animals , Humans , Male , Mice , Diet, High-Fat/adverse effects , Disease Models, Animal , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Mitophagy/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/genetics , Protein Kinases/metabolism , Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...