Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.119
Filter
1.
BMC Urol ; 24(1): 163, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090720

ABSTRACT

BACKGROUND: This study investigated the use of urinary exosomal mRNA as a potential biomarker for the early detection of prostate cancer (PCa). METHODS: Next-generation sequencing was utilized to analyze exosomal RNA from 10 individuals with confirmed PCa and 10 individuals without cancer. Subsequent validation through qRT-PCR in a larger sample of 43 PCa patients and 92 healthy controls revealed distinct mRNA signatures associated with PCa. RESULTS: Notably, mRNAs for RAB5B, WWP1, HIST2H2BF, ZFY, MARK2, PASK, RBM10, and NRSN2 showed promise as diagnostic markers, with AUC values between 0.799 and 0.906 and significance p values. Combining RAB5B and WWP1 in an exoRNA diagnostic model outperformed traditional PSA tests, achieving an AUC of 0.923, 81.4% sensitivity, and 89.1% specificity. CONCLUSIONS: These findings highlight the potential of urinary exosomal mRNA profiling, particularly focusing on RAB5B and WWP1, as a valuable strategy for improving the early detection of PCa.


Subject(s)
Biomarkers, Tumor , Early Detection of Cancer , Exosomes , Prostatic Neoplasms , RNA, Messenger , Humans , Male , Prostatic Neoplasms/urine , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , Exosomes/genetics , RNA, Messenger/urine , Biomarkers, Tumor/urine , Biomarkers, Tumor/genetics , Early Detection of Cancer/methods , Aged , Middle Aged
2.
BMC Cancer ; 24(1): 961, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107726

ABSTRACT

BACKGROUND: Pancreatic cancer (PaCa) is one of the most intractable and fatal malignancies and is associated with the dysregulation of long noncoding RNAs (lncRNAs), which are a large class of noncoding RNAs larger than 200 nt that act as competing endogenous RNAs or sponges for miRNAs to induce tumour biological behaviours. However, their clinical value in treating pancreatic cancer has been poorly explained, but they are essential for improving the prognosis of PaCa patients. METHODS: We analysed the plasma-derived exosomal lncRNA profiles of PaCa patients by using whole-transcriptome sequencing analysis and identified significantly differentially expressed lncRNAs, including LINC01268, LINC02802, AC124854.1, and AL132657.1. In the current study, the expression levels of four plasma-derived exosomal lncRNAs in PaCa plasma were validated via quantitative real-time polymerase chain reaction (qRT‒PCR). The relationship between the expression of the four lncRNAs and the clinicopathological features of patients with PaCa was also evaluated. RESULTS: We demonstrated that exosomal LINC01268, LINC02802, AC124854.1 and AL132657.1 were highly expressed in PaCa plasma compared with those in normal controls; moreover, they were positively correlated with the serum expression of carbohydrate antigen 19-9 (CA19-9). The receiver operating characteristic curves (AUCs) of the four lncRNAs were 0.8421, 0.6544, 0.7190, and 0.6321, and the AUC value of the combination of the four exosomal lncRNAs increased to 0.8476, with a sensitivity of 0.72 and specificity of 0.89. These results suggested that the plasma-derived exosomal genes LINC01268, LINC02802, AC124854.1, and AL132657.1 may be novel diagnostic markers for PaCa. CONCLUSIONS: Our research demonstrated that the plasma-derived exosomal lncRNAs of PaCa patients are novel blood-based biomarkers of disease.


Subject(s)
Biomarkers, Tumor , Exosomes , Pancreatic Neoplasms , RNA, Long Noncoding , Humans , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Exosomes/genetics , Exosomes/metabolism , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Male , Female , Middle Aged , Prognosis , Gene Expression Regulation, Neoplastic , Aged , Gene Expression Profiling/methods , ROC Curve , CA-19-9 Antigen/blood
3.
Clin Transl Med ; 14(8): e1763, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107958

ABSTRACT

BACKGROUND: Breast cancer (BC) is one of the most prevalent malignant tumours that threatens women health worldwide. It has been reported that circular RNAs (circRNAs) play an important role in regulating tumour progression and tumour microenvironment (TME) remodelling. METHODS: Differentially expression characteristics and immune correlations of circRNAs in BC were verified using high-throughput sequencing and bioinformatic analysis. Exosomes were characterised by nanoparticle transmission electron microscopy and tracking analysis. The biological function of circ-0100519 in BC development was demonstrated both in vitro and in vivo. Western blotting, RNA pull-down, RNA immunoprecipitation, flow cytometry, and luciferase reporter were conducted to investigate the underlying mechanism. RESULTS: Circ-0100519 was significant abundant in BC tumour tissues and related to poor prognosis. It can be encapsulated into secreted exosomes, thereby promoting BC cell invasion and metastasis via inducing M2-like macrophages polarisation.Mechanistically, circ-0100519 acted as a scaffold to enhance the interaction between the deubiquitinating enzyme ubiquitin-specific protease 7 (USP7) and nuclear factor-like 2 (NRF2) in macrophages, inducing the USP7-mediated deubiquitination of NRF2. Additionally, HIF-1α could function as an upstream effector to enhance circ-0100519 transcription. CONCLUSIONS: Our study revealed that exosomal circ-0100519 is a potential biomarker for BC diagnosis and prognosis, and the HIF-1α inhibitor PX-478 may provide a therapeutic target for BC.


Subject(s)
Breast Neoplasms , Exosomes , NF-E2-Related Factor 2 , RNA, Circular , Ubiquitin-Specific Peptidase 7 , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Female , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/metabolism , Exosomes/metabolism , Exosomes/genetics , Macrophages/metabolism , Mice , Disease Progression , Animals , Cell Line, Tumor
4.
Sci Rep ; 14(1): 17812, 2024 08 01.
Article in English | MEDLINE | ID: mdl-39090292

ABSTRACT

Chemotherapy, particularly with oxaliplatin, is a key treatment for advanced gastric cancer (GC), and exosomes derived from human bone marrow mesenchymal stem cells (hBM-MSCs) play a vital role in the tumor microenvironment. The study aims to elucidate the previously unexplored role of exosomes derived from hBM-MSCs in GC tumorigenesis, especially under the influence of chemotherapy. We conducted an experimental study, utilizing miRNA sequencing and biological experiments, to analyze the tumorigenicity of exosomal miR-424-3p secreted by hBM-MSCs and its target gene RHOXF2 in GC cell lines. The results were confirmed through experimentation using a xenograft mouse model. This study demonstrated the role of hBM-MSCs in the GC microenvironment, focusing on their epithelial-mesenchymal transition (EMT) facilitation through exosomes, which led to enhanced tumorigenicity in GC cells. Intriguingly, this pro-tumor effect was abrogated when hBM-MSCs were treated with oxaliplatin. Exosomal miRNA sequencing revealed that oxaliplatin can upregulate the levels of miR-424-3p in exosomes secreted by hBM-MSCs, thereby inhibiting the EMT process in GC cells. Furthermore, miR-424-3p was identified to target and downregulate RHOXF2 expression, impeding the malignant behavior of GC cells both in vitro and in the mouse model. These findings uncover a potential hidden mechanism of oxaliplatin's anti-tumor action and propose the delivery of miR-424-3p via exosomes as a promising avenue for anti-tumor therapy.


Subject(s)
Epithelial-Mesenchymal Transition , Exosomes , Mesenchymal Stem Cells , MicroRNAs , Oxaliplatin , Stomach Neoplasms , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Oxaliplatin/pharmacology , Mesenchymal Stem Cells/metabolism , Exosomes/metabolism , Exosomes/genetics , Animals , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Mice , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Up-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Xenograft Model Antitumor Assays , Antineoplastic Agents/pharmacology , Tumor Microenvironment , Mice, Nude , Disease Progression
5.
BMC Cancer ; 24(1): 957, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103776

ABSTRACT

PURPOSE: Exosomal microRNAs have been identified as important mediators of communication between tumor cells and macrophages in the microenvironment. miR-541-5p was reported to be involved in hepatocellular carcinoma progression, but its role in gastric cancer (GC) and in GC cell-macrophage crosstalk is unknown. METHODS: Cell proliferation, migration and invasion were respectively assessed by CCK-8 assay, scratch and Transwell assays. RT-qPCR was used to detect the level of miR-541-5p, macrophage markers and DUSP3. The percentage of CD11b+CD206+ cell population was analyzed by flow cytometry. Western blotting was employed to evaluate DUSP3-JAK2/STAT3 pathway proteins and exosome markers. The interaction between miR-541-5p and DUSP3 was verified by luciferase assay. RESULTS: The results showed that miR-541-5p was upregulated in GC tissues and cells, and stimulated GC cell growth, migration and invasion in vitro. GC cells induce M2 macrophage polarization by secreting the exosomal miR-541-5p. Exosomal miR-541-5p maintained JAK2/STAT3 pathway activation in macrophages by targeting negative regulation of DUSP3. Inhibiting miR-541-5p significantly limited tumor growth in vivo. CONCLUSION: In conclusion, miR-541-5p promotes GC cell progression. GC cells may induce macrophage M2 polarization through the exosomal miR-541-5p-mediated DUSP3/JAK2/STAT3 pathway. miR-541-5p may be a potential therapeutic target for GC.


Subject(s)
Cell Proliferation , Dual Specificity Phosphatase 3 , Exosomes , Janus Kinase 2 , Macrophages , MicroRNAs , STAT3 Transcription Factor , Stomach Neoplasms , Humans , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/metabolism , Exosomes/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Mice , Animals , Macrophages/metabolism , Dual Specificity Phosphatase 3/metabolism , Dual Specificity Phosphatase 3/genetics , Cell Line, Tumor , Signal Transduction , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Male , Female
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(8): 916-924, 2024 Aug 10.
Article in Chinese | MEDLINE | ID: mdl-39097272

ABSTRACT

OBJECTIVE: To explore the clinical phenotype and genetic variant in a Chinese pedigree affected with Hunter syndrome and create immortalized cell lines for the affected pedigree members. METHODS: A pedigree of six members who had visited Xi'an Children's Hospital in July 2022 was selected as the study subject. Clinical data was collected. Whole exome sequencing was carried out for the pedigree members. Candidate variant was verified by Sanger sequencing. In addition, peripheral B lymphocytes were transfected with Epstein-Barr virus to create immortalized cell lines, which were then subjected to enzyme activity analysis. RESULTS: The patient, a five-year-and-seven-month-old boy, had exhibited stiff limbs and enlarged joints. He had developed hernia, scaphocephaly, and barrel chest from 3 months of age. His uncle also had stiff limbs, poor hearing, blindness, and right oblique inguinal hernia. Above features had resembled those of Hunter syndrome. Genetic testing revealed that both the child and his uncle had harbored an IDS (NM_000202.8): c.823G>A (p.D275N) variant, which was unreported previously. Bioinformatic analysis indicated that the D275 to be a highly conserved site, and the D275N variant may affect the stability of the protein's spatial conformation, thereby decrease the catalytic activity of the enzyme. The successfully constructed immortalized lymphoblastoid cell lines for the child and his parents showed increased volume, irregular shape, burr structure and cluster growth. And the value of IDS activity of the patient's immortalized lymphoblastoid cells was below the limit of detection. Based on the guidelines from the American College of Medical Genetics and Genomics (ACMG), the variant was classified as likely pathogenic (PS3+PM2_Supporting+PM5+PP1+PP3). CONCLUSION: Above finding has enriched the phenotypic and mutational spectra of Hunter syndrome, and provided a basis for the genetic counseling for this pedigree. The creation of immortalized cell lines has offered a model for further investigation of the impact of variant on the function of IDS and development of targeted drugs.


Subject(s)
Mucopolysaccharidosis II , Cells, Cultured , Pedigree , Cell Separation , Cell Culture Techniques , Child, Preschool , Mucopolysaccharidosis II/genetics , Genetic Counseling , Female , Phenotype , Hernia/genetics , Exosomes/genetics , Exome Sequencing , Craniosynostoses/genetics , Mutation
7.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125582

ABSTRACT

Human retinal organoids (ROs) have emerged as valuable tools for studying retinal development, modeling human retinal diseases, and screening drugs. However, their application is limited primarily due to time-intensive generation, high costs, and low reproducibility. Quality assessment of RO differentiation is crucial for their application in research. However, traditional methods such as morphological evaluation and immunohistochemical analysis have limitations due to their lack of precision and invasiveness, respectively. This study aims to identify non-invasive biomarkers for RO differentiation quality using exosomal microRNAs (miRNAs), which are known to reflect cell-specific functions and development in the retina. We differentiated ROs from human induced pluripotent stem cells (hiPSCs) and classified them into 'superior' and 'inferior' groups based on morphological and immunohistochemical criteria. Exosomes from the conditioned media were isolated and analyzed for miRNA content. Our findings revealed distinct miRNA profiles between superior and inferior ROs, with superior ROs exhibiting higher miRNA diversity and specifically up- or down-regulated miRNAs. Gene ontology and pathway enrichment analyses indicated that the target genes of these miRNAs are involved in neuron proliferation and differentiation. The study suggests the potential of exosomal hsa-miR-654-3p and hsa-miR-451a as non-invasive biomarkers for real-time monitoring of RO quality, facilitating the development of standardized, efficient, and cost-effective culture methods.


Subject(s)
Biomarkers , Cell Differentiation , Exosomes , Induced Pluripotent Stem Cells , MicroRNAs , Organoids , Retina , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Organoids/metabolism , Organoids/cytology , Cell Differentiation/genetics , Retina/cytology , Retina/metabolism , Biomarkers/metabolism , Exosomes/metabolism , Exosomes/genetics , Cells, Cultured
8.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125689

ABSTRACT

Our study explores the role of cancer-derived extracellular exosomes (EXs), particularly focusing on collagen alpha-3 (VI; COL6A3), in facilitating tumor dissemination and metastasis in epithelial ovarian cancer (EOC). We found that COL6A3 is expressed in aggressive ES2 derivatives, SKOV3 overexpressing COL6A3 (SKOV3/COL6A3), and mesenchymal-type ovarian carcinoma stromal progenitor cells (MSC-OCSPCs), as well as their EXs, but not in less aggressive SKOV3 cells or ES2 cells with COL6A3 knockdown (ES2/shCOL6A3). High COL6A3 expression correlates with worse overall survival among EOC patients, as evidenced by TCGA and GEO data analysis. In vitro experiments showed that EXs from MSC-OCSPCs or SKOV3/COL6A3 cells significantly enhance invasion ability in ES2 or SKOV3/COL6A3 cells, respectively (both, p <0.001). In contrast, ES2 cells with ES2/shCOL6A3 EXs exhibited reduced invasion ability (p < 0.001). In vivo, the average disseminated tumor numbers in the peritoneal cavity were significantly greater in mice receiving intraperitoneally injected SKOV3/COL6A3 cells than in SKOV3 cells (p < 0.001). Furthermore, mice intravenously (IV) injected with SKOV3/COL6A3 cells and SKOV3/COL6A3-EXs showed increased lung colonization compared to mice injected with SKOV3 cells and PBS (p = 0.007) or SKOV3/COL6A3 cells and PBS (p = 0.039). Knockdown of COL6A3 or treatment with EX inhibitor GW4869 or rapamycin-abolished COL6A3-EXs may suppress the aggressiveness of EOC.


Subject(s)
Carcinoma, Ovarian Epithelial , Collagen Type VI , Exosomes , Ovarian Neoplasms , Exosomes/metabolism , Exosomes/genetics , Female , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Humans , Collagen Type VI/metabolism , Collagen Type VI/genetics , Animals , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Mice , Cell Line, Tumor , Neoplasm Metastasis , Neoplasm Invasiveness , Gene Expression Regulation, Neoplastic , Mice, Nude , Cell Movement
9.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125725

ABSTRACT

Specific markers for colorectal cancer (CRC), preceded by colorectal adenoma (pre-CRC), are lacking. This study aimed to investigate whether microRNAs (miR-19a-3p, miR-92a-3p, miR-193a-3p, and miR-210-3p) from tissues and exosomes are potential CRC biomarkers and compare them to existing biomarkers, namely carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9. MiRNA was isolated in the samples of 52 CRC and 76 pre-CRC patients. Expression levels were analyzed by RT-qPCR. When comparing pre-CRC and CRC tissue expression levels, only miR-193a-3p showed statistically significant result (p < 0.0001). When comparing the tissues and exosomes of CRC samples, a statistically significant difference was found for miR-193a-3p (p < 0.0001), miR-19a-3p (p < 0.0001), miR-92a-3p (p = 0.0212), and miR-210-3p (p < 0.0001). A receiver-operating characteristic (ROC) curve and area under the ROC curve (AUC) were used to evaluate the diagnostic value of CEA, CA 19-9, and miRNAs. CEA and CA 19-9 had good diagnostic values (AUCs of 0.798 and 0.668). The diagnostic value only of miR-193a-3p was highlighted (AUC = 0.725). The final logistic regression model, in which we put a combination of CEA concentration and the miR-193a-3p expression level in tissues, showed that using these two markers can distinguish CRC and pre-CRC in 71.3% of cases (AUC = 0.823). MiR-193a-3p from tissues could be a potential CRC biomarker.


Subject(s)
Adenoma , Biomarkers, Tumor , Colorectal Neoplasms , MicroRNAs , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , MicroRNAs/genetics , Biomarkers, Tumor/genetics , Male , Female , Adenoma/genetics , Adenoma/diagnosis , Adenoma/metabolism , Middle Aged , Aged , ROC Curve , Carcinoembryonic Antigen/metabolism , Carcinoembryonic Antigen/genetics , Diagnosis, Differential , Gene Expression Regulation, Neoplastic , Exosomes/genetics , Exosomes/metabolism , Adult , CA-19-9 Antigen , Aged, 80 and over
10.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125827

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 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.


Subject(s)
Exosomes , Mental Disorders , MicroRNAs , Exosomes/metabolism , Exosomes/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Mental Disorders/genetics , Mental Disorders/metabolism , Animals , Stress, Psychological/genetics , Stress, Psychological/metabolism
11.
Int J Mol Med ; 54(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39129308

ABSTRACT

Ovarian cancer (OC) is a common gynecological disease with a high mortality rate worldwide due to its insidious nature and undetectability at an early stage. The standard treatment, combining platinum­based chemotherapy with cytoreductive surgery, has suboptimal results. Therefore, early diagnosis of OC is crucial. All cell types secrete extracellular vesicles, particularly exosomes. Exosomes, which contain lipids, proteins, DNA and non­coding RNAs (ncRNAs), are novel methods of intercellular communication that participate in tumor development and progression. ncRNAs are categorized by size into long ncRNAs (lncRNAs) and small ncRNAs (sncRNAs). sncRNAs further include transfer RNAs, small nucleolar RNAs, PIWI­interacting RNAs and microRNAs (miRNAs). miRNAs inhibit protein translation and promote messenger RNA (mRNA) cleavage to suppress gene expression. By sponging downstream miRNAs, lncRNAs and circular RNAs can regulate target gene expression, thereby weakening the interactions between miRNAs and mRNAs. Exosomes and exosomal ncRNAs, commonly present in human biological fluids, are promising biomarkers for OC. The present article aimed to review the potential role of exosomal ncRNAs in the diagnosis and prognosis of OC by summarizing the characteristics, processes, roles and isolation methods of exosomes and exosomal ncRNAs.


Subject(s)
Exosomes , Ovarian Neoplasms , RNA, Untranslated , Humans , Exosomes/metabolism , Exosomes/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Female , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
12.
Front Immunol ; 15: 1425384, 2024.
Article in English | MEDLINE | ID: mdl-39139565

ABSTRACT

Background: Schistosomiasis is a zoonotic parasitic disorder induced by the infestation of schistosomes, a genus of trematodes. MicroRNAs (miRNAs) in egg-derived exosomes are crucial for modulating the host's immune responses and orchestrating the pathophysiological mechanisms. Although the exosomes secreted by S. japonicum contain abundant miRNAs, the specific roles of these miRNAs in the pathogenesis of schistosomiasis-induced hepatic fibrosis are yet to be comprehensively elucidated. The egg exosomes of S. japonicum secrete miRNA-30, a novel miRNA. Methods: In vitro, the effect of miRNA-30 was evaluated by transfecting HSCs with miRNA mimics. The target gene biosignature for miRNA-30 was predicted using the miRDB software. The effect of miRNA-30 in hepatic fibrosis was evaluated by either elevating its expression in healthy mice or by inhibiting its activity in infected mice by administration of recombinant adeno-associated virus serotype eight vectors expressing miRNA-30 or miRNA sponges. Results: This novel miRNA can activate hepatic stellate cells (HSCs), the prinary effector cells of hepatic fibrosis, in vitro, i.e., it significantly increases the fibrogenic factors Col1(α1), Col3(α1), and α-SMA at both mRNA and protein levels. In addition, miRNA-30 may activate HSCs by targeting the host RORA gene. In addition, in vivo experiments were conducted by administering a recombinant adeno-associated viral vector to modulate the expression levels of miRNA-30. The overexpression of miRNA-30 in healthy mice significantly elevated the expression of Col1(α1), Col3(α1), and α-SMA at both the transcriptomic and proteomic scales. This overexpression was coupled with a pronounced augmentation in the hepatic hydroxyproline content. Conversely, the in vivo silencing of miRNA-30 in infected mice induced a considerable reduction in the size of hepatic granulomas and areas of collagen deposition. Hence, in vivo, modulation of miRNA-30 expression may play a pivotal role in ameliorating the severity of hepatic fibrosis in mice afflicted with S. japonica. Conclusions: The study results suggest that miRNA-30 may augment schistosomiasis-induced hepatic fibrosis through a probable interaction with the host RORA. Our study may improve the current theoretical framework regarding cross-species regulation by miRNAs of hepatic fibrosis in schistosomiasis.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , MicroRNAs , Schistosoma japonicum , Schistosomiasis japonica , Animals , MicroRNAs/genetics , Liver Cirrhosis/parasitology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Schistosomiasis japonica/immunology , Schistosomiasis japonica/genetics , Schistosomiasis japonica/parasitology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/parasitology , Exosomes/metabolism , Exosomes/genetics , Female , Disease Models, Animal , Ovum/metabolism
13.
J Biochem Mol Toxicol ; 38(9): e23769, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39152098

ABSTRACT

Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment (TME) and can induce functional polarization of tumor macrophages. This study aimed to explore the effect of CAFs-derived exosome LINC01833 on the malignant biological behavior of non-small cell lung cancer (NSCLC) cells and its mechanism. Tumor tissues (n = 3) and adjacent noncancerous tissues (n = 3) were collected from patients with NSCLC, and fibroblasts (CAF, NF) were isolated from the two tissues. Expression of LINC01833/miR-335-5p/VAPA in NSCLC clinical tissues and cell lines was detected by RT-qPCR. Exosomes of CAFs and NFs were isolated by ultracentrifugation. Cell proliferation, migration, invasion, and M2 macrophage polarization were detected by MTT, transwell, wound-healing assay, and flow cytometry assay, while western blot was used to verify the expression of M2 macrophage polarization-related proteins. Tumor volume weight and M2 macrophage polarization were detected by tumor xenografts in nude mice. LINC01833 was highly expressed in NSCLC tumor tissues and cells. Knockdown of LINC01833 exosomes could significantly inhibit proliferation, migration, invasion of NSCLC cells, and M2 macrophage polarization of THP-1 cells, while simultaneous knockdown of miR-335-5p on the above basis could reverse the effect of knockdown of LINC01833. In vivo experiments also indicated that knockdown of LINC01833 exosomes suppressed tumor growth and M2 macrophage polarization. CAF-derived LINC01833 exosomes can promote the proliferation, migration and invasion of NSCLC cells and M2 macrophage polarization by inhibiting miR-335-5p and regulating VAPA activity.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Non-Small-Cell Lung , Exosomes , Lung Neoplasms , Mice, Nude , MicroRNAs , RNA, Long Noncoding , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Exosomes/metabolism , Exosomes/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Cell Proliferation , Male , Female , Cell Line, Tumor , Cell Movement , A549 Cells , Mice, Inbred BALB C
14.
Clin Exp Rheumatol ; 42(8): 1629-1634, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39152754

ABSTRACT

OBJECTIVES: To compare the microRNAs (miRNAs) contained within serum exosomes isolated from patients with Raynaud's phenomenon (RP) and negative antinuclear antibodies (ANA) to the miRNA contained in serum exosomes isolated from patients with RP and positive ANA. METHODS: Serum exosomes were isolated employing a polymer precipitation procedure. Next Generation Sequencing (NGS) was used to identify the miRNAs contained in the exosomes isolated from the two clinical cohorts and to analyse the differences in their contents. RESULTS: The NGS results identified six miRNAs that displayed significant differences in their content between serum exosomes from patients with RP with negative serum ANA compared to miRNAs contained in serum exosomes from patients with ANA-positive RP. CONCLUSIONS: A comparative analysis of miRNAs contained within serum exosomes of patients with RP and negative ANA vs. samples from patients with RP and positive ANA identified several differentially expressed miRNAs that may represent non-invasive biomarkers to assist in the identification of patients with RP at risk of evolving into systemic sclerosis.


Subject(s)
Antibodies, Antinuclear , Exosomes , MicroRNAs , Raynaud Disease , Humans , Raynaud Disease/blood , Raynaud Disease/genetics , Raynaud Disease/immunology , Raynaud Disease/diagnosis , Antibodies, Antinuclear/blood , Female , Exosomes/genetics , Middle Aged , MicroRNAs/blood , MicroRNAs/genetics , Male , Adult , Biomarkers/blood , High-Throughput Nucleotide Sequencing , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Aged , Predictive Value of Tests
15.
Biol Direct ; 19(1): 68, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160584

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) have been reported that can affect cancer cell proliferation, metastasis, ferroptosis, and immune escape. METTL3-mediated N6-methyladenine (m6A) modification is involved in the tumorigenesis of colorectal cancer (CRC). Herein, we investigated whether METTL3-dependent m6A in CAFs-derived exosomes (exo) affected CRC progression. METHODS: qRT-PCR and western blotting analyses detected levels of mRNAs and proteins. Cell proliferation and metastasis were evaluated using MTT, colony formation, transwell, and wound healing assays, respectively. Cell ferroptosis was assessed by detecting cell viability and the levels of Fe+, reactive oxygen species, and glutathione after erastin treatment. Exosomes were isolated from CAFs by ultracentrifugation. The m6A modification profile was determined by methylated RNA immunoprecipitation assay and the interaction between METTL3 and ACSL3 (acyl-CoA synthetase 3) was verified using dual-luciferase reporter assay. Animal models were established for in vivo analysis. RESULTS: CAFs promoted CRC cell proliferation and metastasis, and suppressed cell ferroptosis. METTL3 was enriched in CAFs and was packaged into exosomes. The m6A modification and METTL3 expression were increased in CRC samples. Knockdown of METTL3 in CAFs-exo suppressed CRC cell proliferation and metastasis, and induced cell ferroptosis. Mechanistically, METTL3 induced ACSL3 m6A modification and stabilized its expression. The anticancer effects mediated by METTL3-silenced CAFs-exo could be rescued by ACSL3 overexpression. Moreover, in vivo assay also showed that CAFs-exo with decreased METTL3 could hinder CRC growth and metastasis in mice models. CONCLUSION: CAFs promoted the proliferation and metastasis, and restrained the ferroptosis in CRC by exosomal METTL3-elicited ACSL3 m6A modification.


Subject(s)
Cancer-Associated Fibroblasts , Cell Proliferation , Coenzyme A Ligases , Colorectal Neoplasms , Exosomes , Ferroptosis , Methyltransferases , Ferroptosis/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Exosomes/metabolism , Exosomes/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Mice , Cancer-Associated Fibroblasts/metabolism , Animals , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Neoplasm Metastasis , Cell Line, Tumor , Mice, Nude , Male
16.
Mol Cancer ; 23(1): 167, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164756

ABSTRACT

Organs of future metastasis are not passive receivers of circulating tumor cells, but are instead selectively and actively modified by the primary tumor before metastatic spread has even occurred. Tumors orchestrate a pre-metastatic program by conditioning distant organs to create microenvironments that foster the survival and proliferation of tumor cells before their arrival, thereby establishing pre-metastatic niches. Primary tumor-derived exosomes modulate these pre-metastatic niches, generating a permissive environment that facilitates the homing and expansion of tumor cells. Moreover, microRNAs have emerged as a key component of exosomal cargo, serving not only to induce the formation of pre-metastatic niches but also to prime these sites for the arrival and colonization of specific secondary tumor populations. Against this backdrop, this review endeavors to elucidate the impact of tumor-derived exosomal microRNAs on the genesis of their individualized pre-metastatic niches, with a view towards identifying novel means of specifying cancer metastasis and exploiting this phenomenon for cancer immunotherapy.


Subject(s)
Exosomes , MicroRNAs , Neoplasm Metastasis , Neoplasms , Tumor Microenvironment , Humans , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/genetics , Neoplasms/pathology , Neoplasms/genetics , Neoplasms/metabolism , Animals , Gene Expression Regulation, Neoplastic
17.
Mol Cancer ; 23(1): 172, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174949

ABSTRACT

Exosomes mediate cell-to-cell crosstalk involving a variety of biomolecules through an intricate signaling network. In recent years, the pivotal role of exosomes and their non-coding RNAs cargo in the development and progression of several cancer types clearly emerged. In particular, tumor bulk and its microenvironment co-evolve through cellular communications where these nanosized extracellular vesicles are among the most relevant actors. Knowledge about the cellular, and molecular mechanisms involved in these communications will pave the way for novel exosome-based delivery of therapeutic RNAs as well as innovative prognostic/diagnostic tools. Despite the valuable therapeutic potential and clinical relevance of exosomes, their role on sarcoma has been vaguely reported because the rarity and high heterogeneity of this type of cancer. Here, we dissected the scientific literature to unravel the multifaceted role of exosomal non-coding RNAs as mediator of cell-to-cell communications in the sarcoma subtypes.


Subject(s)
Cell Communication , Exosomes , RNA, Untranslated , Sarcoma , Humans , Exosomes/metabolism , Exosomes/genetics , Sarcoma/genetics , Sarcoma/pathology , Sarcoma/therapy , Sarcoma/metabolism , RNA, Untranslated/genetics , Animals , Tumor Microenvironment/genetics , Gene Expression Regulation, Neoplastic , Signal Transduction , Biomarkers, Tumor/genetics , Translational Research, Biomedical
18.
Biomed Pharmacother ; 178: 117231, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094542

ABSTRACT

AIMS: To investigate the molecular mechanism of osteoclast-derived exosomes in osteoporosis. MAIN METHODS: RANKL induced osteoclast model was screened for significantly differentially expressed lncRNAs and mRNAs by whole RNA sequencing. Exosomes were characterized using electron microscopy, western blotting and nanosight. Overexpression or knockdown of AW011738 was performed to explore its function. The degree of osteoporosis in an osteoporosis model was assessed by mirco-CT. The osteoclast model, osteoblast differentiation ability and the molecular mechanism of lncRNA AW011738/miR-24-2-5p/TREM1 axis in osteoporosis were assessed by dual luciferase reporter gene assay, Western blotting (WB), immunofluorescence and ALP staining. Bioinformatics was used to predict interactions of key osteoporosis-related genes with miRNAs, transcription factors, and potential drugs after upregulation of AW011738. To predict the protein-protein interaction (PPI) network associated with key genes, GO and KEGG analyses were performed on the key genes. The ssGSVA was used to predict changes in the immune microenvironment. KEY FINDINGS: Osteoclast-derived exosomes containing lncRNA AW011738 decreased the osteogenesis-related markers and accelerated bone loss in OVX mice. Osteoclast (si-AW011738)-derived exosomes showed a significant increase in biomarkers of osteoblast differentiation in vitro compared to the si-NC group. As analyzed by mirco-CT, tail vein injected si-AW011738 OVX mice were less osteoporotic than the control group. AW011738 inhibited osteoblast differentiation by regulating TREM1 expression through microRNA. Meanwhile, overexpression of miR-24-2-5p inhibited TREM1 expression to promote osteoblast differentiation. SIGNIFICANCE: Osteoclast-derived exosomes containing lncRNA AW011738 inhibit osteogenesis in MC3T3-E1 cells through the lncRNA AW011738/miR-24-2-5p/TREM1 axis and exacerbate osteoporosis in OVX mice.


Subject(s)
Cell Differentiation , Exosomes , MicroRNAs , Osteoblasts , Osteoclasts , Osteoporosis , RNA, Long Noncoding , Triggering Receptor Expressed on Myeloid Cells-1 , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/metabolism , Exosomes/genetics , Osteoblasts/metabolism , Osteoblasts/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Differentiation/genetics , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/metabolism , Mice , Osteoclasts/metabolism , Osteoclasts/pathology , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Female , Osteogenesis/genetics , Mice, Inbred C57BL , Disease Progression
19.
Cell Mol Life Sci ; 81(1): 369, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39182194

ABSTRACT

Calcium-containing stones represent the most common form of kidney calculi, frequently linked to idiopathic hypercalciuria, though their precise pathogenesis remains elusive. This research aimed to elucidate the molecular mechanisms involved by employing urinary exosomal microRNAs as proxies for renal tissue analysis. Elevated miR-148b-5p levels were observed in exosomes derived from patients with kidney stones. Systemic administration of miR-148b-5p in rat models resulted in heightened urinary calcium excretion, whereas its inhibition reduced stone formation. RNA immunoprecipitation combined with deep sequencing identified miR-148b-5p as a suppressor of calcitonin receptor (Calcr) expression, thereby promoting urinary calcium excretion and stone formation. Mice deficient in Calcr in distal epithelial cells demonstrated elevated urinary calcium excretion and renal calcification. Mechanistically, miR-148b-5p regulated Calcr through the circRNA-83536/miR-24-3p signaling pathway. Human kidney tissue samples corroborated these results. In summary, miR-148b-5p regulates the formation of calcium-containing kidney stones via the circRNA-83536/miR-24-3p/Calcr axis, presenting a potential target for novel therapeutic interventions to prevent calcium nephrolithiasis.


Subject(s)
Calcium , Hypercalciuria , MicroRNAs , Nephrolithiasis , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/pathology , Calcium/metabolism , Mice , Rats , Nephrolithiasis/metabolism , Nephrolithiasis/genetics , Nephrolithiasis/pathology , Male , Mice, Inbred C57BL , Kidney Calculi/metabolism , Kidney Calculi/genetics , Rats, Sprague-Dawley , Exosomes/metabolism , Exosomes/genetics , Female , Kidney/metabolism , Kidney/pathology , Mice, Knockout , Signal Transduction
20.
World J Gastroenterol ; 30(22): 2843-2848, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38947286

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common and deadliest subtype of liver cancer worldwide and, therefore, poses an enormous threat to global health. Understanding the molecular mechanisms underlying the development and progression of HCC is central to improving our clinical approaches. PIWI-interacting RNAs (piRNAs) are a class of small non-coding RNAs that bind to PIWI family proteins to regulate gene expression at transcriptional and post-transcriptional levels. A growing body of work shows that the dysregulation of piRNAs plays a crucial role in the progression of various human cancers. In this editorial, we report on the current knowledge of HCC-associated piRNAs and their potential clinical utility. Based on the editorial by Papadopoulos and Trifylli, on the role and clinical evaluation of exosomal circular RNAs in HCC, we highlight this other emerging class of non-coding RNAs.


Subject(s)
Carcinoma, Hepatocellular , Gene Expression Regulation, Neoplastic , Liver Neoplasms , RNA, Small Interfering , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , RNA, Small Interfering/metabolism , Exosomes/metabolism , Exosomes/genetics , RNA, Circular/metabolism , RNA, Circular/genetics , Disease Progression , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL