Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
BMC Pulm Med ; 24(1): 209, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685004

ABSTRACT

BACKGROUND: The pathogenesis of adult non-cystic fibrosis (CF) bronchiectasis is complex, and the relevant molecular mechanism remains ambiguous. Versican (VCAN) is a key factor in inflammation through interactions with adhesion molecules. This study constructs a stable panoramic map of mRNA, reveals the possible pathogenesis of bronchiectasis, and provides new ideas and methods for bronchiectasis. METHODS: Peripheral blood and tissue gene expression data from patients with bronchiectasis and normal control were selected by bioinformatics analysis. The expression of VCAN in peripheral blood and bronchial tissues of bronchiectasis were obtained by transcriptome sequencing. The protein expression levels of VCAN in serums were verified by the enzyme-linked immunosorbent assay (ELISA). The mRNA expression levels of VCAN in co-culture of Pseudomonas aeruginosa and bronchial epithelial cells were verified by real-time quantitative polymerase chain reaction (RT-qPCR). In addition, the biological function of VCAN was detected by the transwell assay. RESULTS: The expression of VCAN was upregulated in the bronchiectasis group by sequencing analysis (P < 0.001). The expression of VCAN in the bronchial epithelial cell line BEAS-2B was increased in P. aeruginosa (P.a), which was co-cultured with BEAS-2B cells (P < 0.05). The concentration of VCAN protein in the serum of patients with bronchiectasis was higher than that in the normal control group (P < 0.05). Transwell experiments showed that exogenous VCAN protein induced the migration of neutrophils (P < 0.0001). CONCLUSIONS: Our findings indicate that VCAN may be involved in the development of bronchiectasis by increasing the migration of neutrophils and play an important role in bronchial pathogenesis.


Subject(s)
Bronchiectasis , Versicans , Humans , Male , Female , Middle Aged , Retrospective Studies , Versicans/genetics , Versicans/metabolism , Adult , Pseudomonas aeruginosa/genetics , Epithelial Cells/metabolism , Aged , Up-Regulation , Coculture Techniques , Bronchi/pathology , Cell Line , RNA, Messenger/metabolism , Case-Control Studies , Clinical Relevance
2.
Circulation ; 149(13): 1004-1015, 2024 03 26.
Article in English | MEDLINE | ID: mdl-37886839

ABSTRACT

BACKGROUND: The adult mammalian heart is incapable of regeneration, whereas a transient regenerative capacity is maintained in the neonatal heart, primarily through the proliferation of preexisting cardiomyocytes. Neonatal heart regeneration after myocardial injury is accompanied by an expansion of cardiac fibroblasts and compositional changes in the extracellular matrix. Whether and how these changes influence cardiomyocyte proliferation and heart regeneration remains to be investigated. METHODS: We used apical resection and myocardial infarction surgical models in neonatal and adult mice to investigate extracellular matrix components involved in heart regeneration after injury. Single-cell RNA sequencing and liquid chromatography-mass spectrometry analyses were used for versican identification. Cardiac fibroblast-specific Vcan deletion was achieved using the mouse strains Col1a2-2A-CreER and Vcanfl/fl. Molecular signaling pathways related to the effects of versican were assessed through Western blot, immunostaining, and quantitative reverse transcription polymerase chain reaction. Cardiac fibrosis and heart function were evaluated by Masson trichrome staining and echocardiography, respectively. RESULTS: Versican, a cardiac fibroblast-derived extracellular matrix component, was upregulated after neonatal myocardial injury and promoted cardiomyocyte proliferation. Conditional knockout of Vcan in cardiac fibroblasts decreased cardiomyocyte proliferation and impaired neonatal heart regeneration. In adult mice, intramyocardial injection of versican after myocardial infarction enhanced cardiomyocyte proliferation, reduced fibrosis, and improved cardiac function. Furthermore, versican augmented the proliferation of human induced pluripotent stem cell-derived cardiomyocytes. Mechanistically, versican activated integrin ß1 and downstream signaling molecules, including ERK1/2 and Akt, thereby promoting cardiomyocyte proliferation and cardiac repair. CONCLUSIONS: Our study identifies versican as a cardiac fibroblast-derived pro-proliferative proteoglycan and clarifies the role of versican in promoting adult cardiac repair. These findings highlight its potential as a therapeutic factor for ischemic heart diseases.


Subject(s)
Heart Injuries , Induced Pluripotent Stem Cells , Myocardial Infarction , Animals , Humans , Mice , Animals, Newborn , Cell Proliferation , Heart , Heart Injuries/metabolism , Induced Pluripotent Stem Cells/metabolism , Mammals , Myocytes, Cardiac/metabolism , Regeneration , Versicans/genetics , Versicans/metabolism
3.
Redox Biol ; 64: 102794, 2023 08.
Article in English | MEDLINE | ID: mdl-37402332

ABSTRACT

Continued oxidant production during chronic inflammation generates host tissue damage, with this being associated with pathologies including atherosclerosis. Atherosclerotic plaques contain modified proteins that may contribute to disease development, including plaque rupture, the major cause of heart attacks and strokes. Versican, a large extracellular matrix (ECM) chondroitin-sulfate proteoglycan, accumulates during atherogenesis, where it interacts with other ECM proteins, receptors and hyaluronan, and promotes inflammation. As activated leukocytes produce oxidants including peroxynitrite/peroxynitrous acid (ONOO-/ONOOH) at sites of inflammation, we hypothesized that versican is an oxidant target, with this resulting in structural and functional changes that may exacerbate plaque development. The recombinant human V3 isoform of versican becomes aggregated on exposure to ONOO-/ONOOH. Both reagent ONOO-/ONOOH and SIN-1 (a thermal source of ONOO-/ONOOH) modified Tyr, Trp and Met residues. ONOO-/ONOOH mainly favors nitration of Tyr, whereas SIN-1 mostly induced hydroxylation of Tyr, and oxidation of Trp and Met. Peptide mass mapping indicated 26 sites with modifications (15 Tyr, 5 Trp, 6 Met), with the extent of modification quantified at 16. Multiple modifications, including the most extensively nitrated residue (Tyr161), are within the hyaluronan-binding region, and associated with decreased hyaluronan binding. ONOO-/ONOOH modification also resulted in decreased cell adhesion and increased proliferation of human coronary artery smooth muscle cells. Evidence is also presented for colocalization of versican and 3-nitrotyrosine epitopes in advanced (type II-III) human atherosclerotic plaques. In conclusion, versican is readily modified by ONOO-/ONOOH, resulting in chemical and structural modifications that affect protein function, including hyaluronan binding and cell interactions.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Oxidants/metabolism , Peroxynitrous Acid/metabolism , Versicans/genetics , Versicans/metabolism , Hyaluronic Acid/metabolism , Plaque, Atherosclerotic/metabolism , Extracellular Matrix/metabolism , Atherosclerosis/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Inflammation/metabolism
4.
Am J Physiol Cell Physiol ; 325(2): C519-C537, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37399500

ABSTRACT

V3 is an isoform of the extracellular matrix (ECM) proteoglycan (PG) versican generated through alternative splicing of the versican gene such that the two major exons coding for sequences in the protein core that support chondroitin sulfate (CS) glycosaminoglycan (GAG) chain attachment are excluded. Thus, versican V3 isoform carries no GAGs. A survey of PubMed reveals only 50 publications specifically on V3 versican, so it is a very understudied member of the versican family, partly because to date there are no antibodies that can distinguish V3 from the CS-carrying isoforms of versican, that is, to facilitate functional and mechanistic studies. However, a number of in vitro and in vivo studies have identified the expression of the V3 transcript during different phases of development and in disease, and selective overexpression of V3 has shown dramatic phenotypic effects in "gain and loss of function" studies in experimental models. Thus, we thought it would be useful and instructive to discuss the discovery, characterization, and the putative biological importance of the enigmatic V3 isoform of versican.


Subject(s)
Alternative Splicing , Versicans , Extracellular Matrix , Protein Isoforms/genetics , Versicans/genetics , Humans
5.
Molecules ; 28(9)2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37175152

ABSTRACT

Versican is a chondroitin sulfate proteoglycan (CSPG), which deposits in perineurium as a physical barrier and prevents the growth of axons out of the fascial boundary. Several studies have indicated that the chondroitin sulfate (CS) chains on versican have several possible functions beyond the physical barrier, including the ability to stabilize versican core protein in the extracellular matrix. As chondroitin sulfate synthase 1 (Chsy1) is a crucial enzyme for CS elongation, we hypothesized that in vivo knockdown of Chsy1 at peripheral nerve lesion site may decrease CS and versican accumulation, and result in accelerating neurite regeneration. In the present study, end-to-side neurorrhaphy (ESN) in Wistar rats was used as an in vivo model of peripheral nerve injury to evaluate nerve regeneration after surgical intervention. The distribution and expression of versican and Chsy1 in regenerating axons after ESN was studied using confocal microscopy and western blotting. Chsy1 was silenced at the nerve lesion (surgical) site using in vivo siRNA transfection. The results indicated that Chsy1 was successfully silenced in nerve tissue, and its downregulation was associated with functional recovery of compound muscle action potential. Silencing of Chsy1 also decreased the accumulation of versican core protein, suggesting that transient treating of Chsy1-siRNA may be an alternative and an effective strategy to promote injured peripheral nerve regeneration.


Subject(s)
Chondroitin Sulfates , Versicans , Rats , Animals , Versicans/genetics , Chondroitin Sulfates/pharmacology , Rats, Wistar , Axons/metabolism , Nerve Regeneration , RNA, Small Interfering/pharmacology
6.
Int J Mol Sci ; 24(8)2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37108649

ABSTRACT

Versican (VCAN), also known as extracellular matrix proteoglycan 2, has been suggested as a potential biomarker in cancers. Previous research has found that VCAN is highly expressed in bladder cancer. However, its role in predicting outcomes for patients with upper urinary tract urothelial cancer (UTUC) is not well understood. In this study, we collected tissues from 10 patients with UTUC, including 6 with and 4 without lymphovascular invasion (LVI), a pathological feature that plays a significant role in determining metastasis. Results from RNA sequencing revealed that the most differentially expressed genes were involved in extracellular matrix organization. Using the TCGA database for clinical correlation, VCAN was identified as a target for study. A chromosome methylation assay showed that VCAN was hypomethylated in tumors with LVI. In our patient samples, VCAN expression was also found to be high in UTUC tumors with LVI. In vitro analysis showed that knocking down VCAN inhibited cell migration but not proliferation. A heatmap analysis also confirmed a significant correlation between VCAN and migration genes. Additionally, silencing VCAN increased the effectiveness of cisplatin, gemcitabine and epirubicin, thus providing potential opportunities for clinical application.


Subject(s)
Carcinoma, Transitional Cell , Kidney Neoplasms , Urinary Bladder Neoplasms , Urinary Tract , Humans , Carcinoma, Transitional Cell/pathology , Versicans/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Kidney Neoplasms/pathology , Biomarkers, Tumor/genetics , Urinary Tract/pathology
7.
Appl Biochem Biotechnol ; 195(12): 7568-7582, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37032373

ABSTRACT

Previous research indicated that the dysregulation of miRNA-30a-5p has a correlation with cell metastasis of lung adenocarcinoma (LUAD). But the study about the molecular regulatory mechanism of miRNA-30a-5p in LUAD cell metastasis is limited. Thus, we discussed the mechanism of miRNA-30a-5p and its biological function in LUAD cells. By utilizing bioinformatics analysis, how miRNA-30a-5p was expressed in LUAD tissue was determined and its downstream target genes were predicted. The signaling pathways where these target genes enriched were analyzed. Several in vitro experiments were applied for cell function detection: dual-luciferase assay for validating the targeting relationship between miRNA-30a-5p and its target gene; quantitative real-time polymerase chain reaction for testing the expression of miRNA-30a-5p and its target gene in LUAD cells; MTT, transwell, cell adhesion, flow cytometry and immunofluorescence assays for examining the capabilities of LUAD cells to proliferate, migrate, invade, adhere, apoptosis and epithelial-mesenchymal transition (EMT) effect; Western blot for determining the expression of adhesion-related proteins and EMT-related proteins. Down-regulated miRNA-30a-5p was discovered in LUAD cells, but on the contrary, VCAN was upregulated. MiRNA-30a-5p overexpression notably repressed the virulent progression of LUAD cells. Besides, dual-luciferase assay validated the targeting relationship between miRNA-30a-5p and VCAN. MiRNA-30a-5p, by negatively regulating VCAN, was capable of hindering LUAD cell proliferation, migration, invasion, adhesion, viability and EMT. It was illustrated that miRNA-30a-5p could downregulate VCAN to retard the malignant progression of LUAD cells, which provides novel insights into LUAD pathogenesis, suggesting that miRNA-30a-5p/VCAN axis can be a promising anti-cancer target for LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Lung Neoplasms/pathology , Cell Proliferation/genetics , Luciferases/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Versicans/genetics , Versicans/metabolism
8.
Medicine (Baltimore) ; 102(11): e33299, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36930067

ABSTRACT

BACKGROUND: Biliary atresia (BA) is a devastating progressive fibro inflammatory disorder in infants. The exact etiology of BA is still unclear. This study aimed screen key genes potentially associated with the occurrence of BA. METHODS: All BA data was obtained from GSE46960 dataset. The limma package in R language was used for differentially expressed gene (DEG) analyses. gene ontology and Kyoto encyclopedia of genes and genomes enrichment analysis were performed on the screened DEGs, using "clusterProfiler" package. protein-protein interaction network was built based on STRING Cytoscape software (Bethesda, Rockville, MD). The logistic regression model was constructed based on the selected DEGs. RESULTS: There were totally 78 DEGs in BA samples compared with normal samples, which were significantly enriched in 200 biological process terms, 37 molecular function terms, 17 cellular component terms, and 18 Kyoto encyclopedia of genes and genomes pathways. Among which, the top 10 genes with the highest importance in protein-protein interaction network were selected. Subsequently, on the basis of the stepwise regression method and 5-fold cross-validation, the logistic regression model constructed based on COL3A1, CXCL8, VCAN, THBS2, and COL1A2 was finally evidenced to predict the BA sample relatively reliably. CONCLUSIONS: In conclusion, COL3A1, CXCL8, VCAN, THBS2, and COL1A2 are potentially crucial genes in BA. The logistic regression model constructed based on them could predict the BA sample relatively reliably.


Subject(s)
Biliary Atresia , Gene Expression Profiling , Humans , Biliary Atresia/genetics , Collagen Type I/genetics , Collagen Type III , Computational Biology/methods , Protein Interaction Maps/genetics , Versicans/genetics
9.
Int J Mol Sci ; 24(6)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36982775

ABSTRACT

Androgenic alopecia (AGA) is the most common type of hair loss, where local high concentrations of dihydrotestosterone (DHT) in the scalp cause progressive shrinkage of the hair follicles, eventually contributing to hair loss. Due to the limitations of existing methods to treat AGA, the use of multi-origin mesenchymal stromal cell-derived exosomes has been proposed. However, the functions and mechanisms of action of exosomes secreted by adipose mesenchymal stromal cells (ADSCs-Exos) in AGA are still unclear. Using Cell Counting Kit-8 (CCK8) analysis, immunofluorescence staining, scratch assays, and Western blotting, it was found that ADSC-Exos contributed to the proliferation, migration, and differentiation of dermal papilla cells (DPCs) and up-regulated the expression of cyclin, ß-catenin, versican, and BMP2. ADSC-Exos also mitigated the inhibitory effects of DHT on DPCs and down-regulated transforming growth factor-beta1 (TGF-ß1) and its downstream genes. Moreover, high-throughput miRNA sequencing and bioinformatics analysis identified 225 genes that were co-expressed in ADSC-Exos; of these, miR-122-5p was highly enriched and was found by luciferase assays to target SMAD3. ADSC-Exos carrying miR-122-5p antagonized DHT inhibition of hair follicles, up-regulated the expression of ß-catenin and versican in vivo and in vitro, restored hair bulb size and dermal thickness, and promoted the normal growth of hair follicles. So, ADSC-Exos enhanced the regeneration of hair follicles in AGA through the action of miR-122-5p and the inhibition of the TGF-ß/SMAD3 axis. These results suggest a novel treatment option for the treatment of AGA.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Humans , Hair Follicle/metabolism , Transforming Growth Factor beta1/metabolism , Dihydrotestosterone/pharmacology , Dihydrotestosterone/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Exosomes/metabolism , Versicans/genetics , Versicans/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction , MicroRNAs/genetics , MicroRNAs/metabolism , Alopecia/metabolism , Smad3 Protein/metabolism
10.
Aging (Albany NY) ; 15(5): 1343-1357, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36842141

ABSTRACT

OBJECTIVE: Gastric cancer is the most common malignant tumor of the digestive system. The progression from gastritis to gastric cancer may be related to genetic factors, but the specific molecular mechanism remains unclear. Therefore, an in-depth study of the molecular mechanism of gastritis and gastric cancer is significant. METHODS: We downloaded two gene profiles, GSE2669 and GSE116312, from the Gene Expression Omnibus (GEO) database. This study aims to apply bioinformatics technology to mine differentially expressed genes (DEGs), DEGs annotation, protein-protein interaction (PPI) network creation, and hub gene identification and expression between gastric cancer patients and gastritis patients. Overall survival analysis of hub genes, analysis by comparative toxicogenomics database for hub genes in gastric cancer, THBS2 and VCAN protein expression by immunohistochemistry for gastric cancer and gastritis as well as design of the biological process (BP) neural network was implemented. RESULTS: The MSLN, SPP1, THBS2, SPARC, FN1, IGFBP7, VCAN were up-regulated in gastric carcinoma samples, while FGA was down-regulated. The protein expression of THBS2 and VCAN in gastric cancer was significantly higher than that in gastritis. VCAN protein expression was positively associated with tumor invasion (P = 0.011) and HER2 overexpression (P = 0.031). Strong correlation among THBS2, VCAN, and gastric cancer based on the BP neural network. CONCLUSION: THBS2 and VCAN may be potential targets for improving gastric cancer patients' diagnosis and clinical efficacy.


Subject(s)
Gene Expression Profiling , Stomach Neoplasms , Humans , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Computational Biology , Prognosis , Protein Interaction Maps/genetics , Stomach Neoplasms/pathology , Versicans/genetics
11.
Biochem Biophys Res Commun ; 643: 175-185, 2023 02 05.
Article in English | MEDLINE | ID: mdl-36621113

ABSTRACT

BACKGROUND: Thoracic aortic aneurysm (TAA) is a silent but dangerous cardiovascular disease. Understanding molecular mechanisms of TAA on single-cell level might provide new strategies for preventing and treating TAA. METHODS: Single-cell RNA sequencing was performed on control and aneurysmal thoracic aorta to find out specific cell clusters and cell types. Western blot and histological staining were used to verify the findings of single-cell transcriptome analysis. Characteristics of Versican (VCAN) overexpressed myofibroblast was evaluated through bioinformatic methods and experimental validation. RESULTS: A total of 3 control and 8 TAA specimens were used for single-cell transcriptome analysis including 48,128 thoracic aortic cells. Among these cells, we found out a specific cell cluster containing both hallmarks of smooth muscle cell (SMC) and fibroblast. Thus, we defined these cells as myofibroblast. Further single-cell transcriptome analysis identified VCAN as a cellular marker of myofibroblast. Western blot and histological staining revealed that VCAN(+) myofibroblast was significantly increased in TAA specimens compared with control individuals. Differential analysis, functional, pathway enrichment analysis and cell-cell communication analysis demonstrated that VCAN(+) myofibroblast was closely associated with previous reported TAA associated pathological process including SMC proliferation, SMC migration and extracellular matrix (ECM) disruption. Pathway analysis found out significant activation of PI3K-AKT signaling pathway within VCAN(+) myofibroblast, which was further confirmed by experimental validation. CONCLUSIONS: Single-cell RNA sequencing identified VCAN(+) myofibroblast as a typical cellular hallmark of TAA. These cells might participate in the pathogenesis of TAA through activation of PI3K-AKT signaling pathway to link SMC proliferation, SMC migration and ECM disruption.


Subject(s)
Aortic Aneurysm, Thoracic , Versicans , Humans , Versicans/genetics , Versicans/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Myofibroblasts/metabolism , Single-Cell Gene Expression Analysis , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aorta, Thoracic/metabolism , Signal Transduction
12.
Matrix Biol ; 115: 16-31, 2023 01.
Article in English | MEDLINE | ID: mdl-36423736

ABSTRACT

The extracellular matrix (ECM) in the endometrium plays a crucial role in mammalian pregnancy. We have shown that versican secreted from the endometrial epithelium promotes embryo implantation. Versican is a proteoglycan, a major player in the provisional matrix, and versikine, its N-terminal fragment cleaved by ADAMTS proteinases, serves as a bioactive molecule. Here, since versican expression in the placenta was dynamically altered in humans and mice, we investigated the role of versican in pregnancy using uterine-specific Vcan deletion mice (uKO mice) and ADAMTS-resistant versican expressing mice (V1R mice). uKO mice exhibited insufficient spiral artery dilation, followed by fetal growth restriction and maternal hypertension. Further analysis revealed impaired proliferation of tissue-resident natural killer cells required for spiral artery dilation. V1R mice showed the same results as the control, eliminating the involvement of versikine. Our results provide a new concept that versican, one factor of ECM, contributes to placentation and following fetal growth.


Subject(s)
Uterus , Versicans , Pregnancy , Humans , Female , Mice , Animals , Versicans/genetics , Versicans/metabolism , Dilatation , Uterus/metabolism , Fetal Development , Arteries/metabolism , Mammals/metabolism
13.
Mol Genet Genomic Med ; 11(2): e2083, 2023 02.
Article in English | MEDLINE | ID: mdl-36333947

ABSTRACT

BACKGROUND: Wagner vitreoretinopathy (WVR) is a rare autosomal dominant vitreoretinopathy caused by pathogenic variants in the VCAN gene. The aim of this study was to report a novel splicing variant in VCAN identified in a three-generation Chinese family initially diagnosed with familial exudative vitreoretinopathy and to describe the patients' clinical features. METHODS: Four affected individuals from a three-generation family underwent detailed ophthalmic examinations, including best-corrected visual acuity by Snellen E chart, slit-lamp biomicroscopy, indirect ophthalmoscopy under pupil dilatation, ocular B-ultrasonography, optical coherence tomography scans, and fundus autofluorescence. Targeted next-generation sequencing was performed to identify variants of the disease-causing gene for the proband, followed by co-segregation analysis using Sanger-DNA sequencing. Reverse transcriptase-polymerase chain reaction (RT-PCR) was carried out to verify the effects of a variant on VCAN pre-mRNA splicing in the lymphocytes from the patients. RESULTS: We detected a novel heterozygous variant c.4004-4_c.4004-3delinsCA of VCAN in all four affected individuals. RT-PCR revealed that the novel variant caused an abnormal splicing in exon 8 of the VCAN and imbalanced versican transcripts. All four patients presented vitreous syneresis and bilateral retinal detachment occurring at different ages. The patients also showed different extents of visual defects and diverse clinical manifestations, including cataract, iris-lens synechiae, inverted papillae, and ectopic foveas. CONCLUSIONS: Our results expand the mutation spectrum of VCAN and further confirm that the splicing sites for exon 8 are mutation hot spots. Patients with WVR may present high phenotype variation; therefore, molecular analysis is very important for precise diagnosis of patients with inherited vitreoretinopathy.


Subject(s)
East Asian People , Retinal Degeneration , Versicans , Humans , Familial Exudative Vitreoretinopathies/genetics , Pedigree , Retina/pathology , Retinal Degeneration/diagnosis , Versicans/genetics
14.
Front Immunol ; 13: 960570, 2022.
Article in English | MEDLINE | ID: mdl-36203562

ABSTRACT

Background: Increasing evidence has revealed an important role of versican (VCAN) on various aspects of cancer progression. Here, we assessed the impact of VCAN expression on prognosis and the response to adjuvant therapy and immunotherapy in patients with gastric cancer (GC). Methods: Four independent cohorts containing 1353 patients with GC, were utilized to investigate the effect of VCAN expression on prognosis and response to adjuvant therapy in GC. Two cohorts treated with immune checkpoint blockades were included to assess the predict value of VCAN expression on response to immunotherapy. Moreover, the bulk RNA-seq and single-cell RNA-seq data were analyzed to illustrate the role of VCAN in tumor microenvironment. Clinical outcomes of patient subgroups were compared by Kaplan-Meier curves with the log-rank test. Result: High VCAN expression was associated with poor prognosis for patients with GC. Compared with patients with high VCAN expression, patients with low VCAN expression benefited more from adjuvant chemotherapy and adjuvant chemoradiotherapy. Moreover, patients with high VCAN expression tended to be resistant to immunotherapy, and VCAN could serve as a promising indicator for predicting the response to immunotherapy. VCANhigh tumors showed a specific microenvironment with more cancer associated fibroblasts infiltration and significant enrichment of stromal relevant signaling pathways. Conclusion: VCAN could predict the response to adjuvant chemotherapy, adjuvant chemoradiotherapy and immunotherapy in GC, and designing new medicine target to VCAN might be an effective way to improve the efficacy of several treatment options for GC.


Subject(s)
Stomach Neoplasms , Versicans , Humans , Immunotherapy , Prognosis , Stomach Neoplasms/therapy , Tumor Microenvironment , Versicans/genetics
15.
Biomolecules ; 12(9)2022 09 13.
Article in English | MEDLINE | ID: mdl-36139132

ABSTRACT

In pigs, meat quality and production are two important traits affecting the pig industry and human health. Compared to lean-type pigs, fat-type pigs contain higher intramuscular fat (IMF) contents, better taste and nutritional value. To uncover genetic factors controlling differences related to IMF in pig muscle, we performed RNA-seq analysis on the transcriptomes of the Longissimus dorsi (LD) muscle of Laiwu pigs (LW, fat-type pigs) and commercial Duroc × Landrace × Yorkshire pigs (DLY, lean-type pigs) at 150 d to compare the expression profiles of mRNA, miRNA and lncRNA. A total of 225 mRNAs, 12 miRNAs and 57 lncRNAs were found to be differentially expressed at the criteria of |log2(foldchange)| > 1 and q < 0.05. The mRNA expression of LDHB was significantly higher in the LD muscle of LW compared to DLY pigs with log2(foldchange) being 9.66. Using protein interaction prediction method, we identified more interactions of estrogen-related receptor alpha (ESRRA) associated with upregulated mRNAs, whereas versican (VCAN) and proenkephalin (PENK) were associated with downregulated mRNAs in LW pigs. Integrated analysis on differentially expressed (DE) mRNAs and miRNAs in the LD muscle between LW and DLY pigs revealed two network modules: between five upregulated mRNA genes (GALNT15, FKBP5, PPARGC1A, LOC110258214 and LOC110258215) and six downregulated miRNA genes (ssc-let-7a, ssc-miR190-3p, ssc-miR356-5p, ssc-miR573-5p, ssc-miR204-5p and ssc-miR-10383), and between three downregulated DE mRNA genes (IFRD1, LOC110258600 and LOC102158401) and six upregulated DE miRNA genes (ssc-miR1379-3p, ssc-miR1379-5p, ssc-miR397-5p, ssc-miR1358-5p, ssc-miR299-5p and ssc-miR1156-5p) in LW pigs. Based on the mRNA and ncRNA binding site targeting database, we constructed a regulatory network with miRNA as the center and mRNA and lncRNA as the target genes, including GALNT15/ssc-let-7a/LOC100523888, IFRD1/ssc-miR1379-5p/CD99, etc., forming a ceRNA network in the LD muscles that are differentially expressed between LW and DLY pigs. Collectively, these data may provide resources for further investigation of molecular mechanisms underlying differences in meat traits between lean- and fat-type pigs.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Muscles/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Swine/genetics , Transcriptome , Versicans/genetics
16.
Molecules ; 27(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36144730

ABSTRACT

We analyzed the expression of ADAMTS proteinases ADAMTS-1, -2, -4, -5 and -13; their activating enzyme MMP-15; and the degradation products of proteoglycan substrates versican and biglycan in an ocular microenvironment of proliferative diabetic retinopathy (PDR) patients. Vitreous samples from PDR and nondiabetic patients, epiretinal fibrovascular membranes from PDR patients, rat retinas, retinal Müller glial cells and human retinal microvascular endothelial cells (HRMECs) were studied. The levels of ADAMTS proteinases and MMP-15 were increased in the vitreous from PDR patients. Both full-length and cleaved activation/degradation fragments of ADAMTS proteinases were identified. The amounts of versican and biglycan cleavage products were increased in vitreous from PDR patients. ADAMTS proteinases and MMP-15 were localized in endothelial cells, monocytes/macrophages and myofibroblasts in PDR membranes, and ADAMTS-4 was expressed in the highest number of stromal cells. The angiogenic activity of PDR membranes correlated significantly with levels of ADAMTS-1 and -4 cellular expression. ADAMTS proteinases and MMP-15 were expressed in rat retinas. ADAMTS-1 and -5 and MMP-15 levels were increased in diabetic rat retinas. HRMECs and Müller cells constitutively expressed ADAMTS proteinases but not MMP-15. The inhibition of NF-κB significantly attenuated the TNF-α-and-VEGF-induced upregulation of ADAMTS-1 and -4 in a culture medium of HRMECs and Müller cells. In conclusion, ADAMTS proteinases, MMP-15 and versican and biglycan cleavage products were increased in the ocular microenvironment of patients with PDR.


Subject(s)
ADAMTS Proteins/metabolism , Diabetes Mellitus, Experimental , Diabetic Retinopathy , Animals , Biglycan/metabolism , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , NF-kappa B/metabolism , Peptide Hydrolases/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Versicans/genetics , Versicans/metabolism , Vitreous Body/metabolism
17.
Article in English | MEDLINE | ID: mdl-36041709

ABSTRACT

Ovarian cyclicity is variable in adult Siberian hamsters (Phodopus sungorus), who respond to long breeding season photoperiods with follicle development and ovulation, while short photoperiods typical of the non-breeding season induce gonadal atrophy. Recent RNAseq results identified ovarian matrix components and regulators of metabolism as differentially regulated by photoperiod; however, the impact of photoperiod across a full cycle of ovarian regression and recrudescence had not been explored for additional regulators of ovarian metabolism and extracellular matrix components. We hypothesized that matrix and metabolism-related genes would be expressed differentially across photoperiods that mimic breeding and non-breeding season daylengths. Hamsters were housed in one of four photoperiod groups: long day (16 h of light per day: 8 h of dark; LD, controls), short day regressed (8 L:16D; SD, regressed), and females exposed to SD then transferred to LD to stimulate return of ovarian function for 2 (early recrudescence), or 8 (late recrudescence) weeks. Plasma leptin concentrations along with expression of ovarian versican and liver-receptor homolog-1/Nr582 mRNA decreased in SD compared to LD and late recrudescence, while vimentin mRNA expression peaked in early and late recrudescence. Ovarian expression of fibronectin and extracellular matrix protein-1 was low in LD ovaries and increased in regressed and recrudescing groups. Expression of hyaluronidase-2, nectin-2, liver-X receptors-α and-ß, and adiponectin mRNA peaked in late recrudescence, with no changes noted for adiponectin receptor-1 and -2. The results offer a first look at the parallels between expression of these genes and the dynamic remodeling that occurs during ovarian regression and recrudescence.


Subject(s)
Ovary , Phodopus , Adiponectin/genetics , Adiponectin/metabolism , Animals , Cricetinae , Extracellular Matrix/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism , Leptin/metabolism , Nectins/genetics , Nectins/metabolism , Ovary/metabolism , Phodopus/physiology , Photoperiod , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Recurrence , Seasons , Versicans/genetics , Versicans/metabolism , Vimentin/genetics , Vimentin/metabolism
18.
Am J Physiol Cell Physiol ; 323(2): C249-C276, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35649251

ABSTRACT

The extracellular matrix (ECM) imparts critical mechanical and biochemical information to cells in the lungs. Proteoglycans are essential constituents of the ECM and play a crucial role in controlling numerous biological processes, including regulating cellular phenotype and function. Versican, a chondroitin sulfate proteoglycan required for embryonic development, is almost absent from mature, healthy lungs and is reexpressed and accumulates in acute and chronic lung disease. Studies using genetically engineered mice show that the versican-enriched matrix can be pro- or anti-inflammatory depending on the cellular source or disease process studied. The mechanisms whereby versican develops a contextual ECM remain largely unknown. The primary goal of this review is to provide an overview of the interaction of versican with its many binding partners, the "versican interactome," and how through these interactions, versican is an integrator of complex extracellular information. Hopefully, the information provided in this review will be used to develop future studies to determine how versican and its binding partners can develop contextual ECMs that control select biological processes. Although this review focuses on versican and the lungs, what is described can be extended to other proteoglycans, tissues, and organs.


Subject(s)
Extracellular Matrix , Versicans , Animals , Extracellular Matrix/metabolism , Lung/metabolism , Mice , Versicans/genetics , Versicans/metabolism
19.
Cancer Lett ; 543: 215779, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35697329

ABSTRACT

High grade serous ovarian cancers (HGSOC) predominantly arise in the fallopian tube epithelium (FTE) and colonize the ovary first, before further metastasis to the peritoneum. Ovarian cancer risk is directly related to the number of ovulations, suggesting that the ovary may secrete specific factors that act as chemoattractants for fallopian tube derived tumor cells during ovulation. We found that 3D ovarian organ culture produced a secreted factor that enhanced the migration of FTE non-tumorigenic cells as well as cells harboring specific pathway modifications commonly found in high grade serous cancers. Through size fractionation and a small molecule inhibitors screen, the secreted protein was determined to be 50-100kDa in size and acted through the Epidermal Growth Factor Receptor (EGFR). To correlate the candidates with ovulation, the PREDICT organ-on-chip system was optimized to support ovulation in a perfused microfluidic platform. Versican was found in the correct molecular weight range, contained EGF-like domains, and correlated with ovulation in the PREDICT system. Exogenous versican increased migration, invasion, and enhanced adhesion of both murine and human FTE cells to the ovary in an EGFR-dependent manner. The identification of a protein secreted during ovulation that impacts the ability of FTE cells to colonize the ovary provides new insights into the development of strategies for limiting primary ovarian metastasis.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Animals , Cystadenocarcinoma, Serous/pathology , ErbB Receptors , Fallopian Tube Neoplasms/pathology , Fallopian Tubes/pathology , Female , Humans , Mice , Ovarian Neoplasms/pathology , Ovulation , Versicans/genetics
20.
Sci Rep ; 12(1): 7282, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508614

ABSTRACT

As photoreceptor cells die during retinal degeneration, the surrounding microenvironment undergoes significant changes that are increasingly recognized to play a prominent role in determining the efficacy of therapeutic interventions. Chondroitin Sulphate Proteoglycans (CSPGs) are a major component of the extracellular matrix that have been shown to inhibit neuronal regrowth and regeneration in the brain and spinal cord, but comparatively little is known about their expression in retinal degeneration. Here we provide a comprehensive atlas of the expression patterns of four individual CSPGs in three models of inherited retinal degeneration and wildtype mice. In wildtype mice, Aggrecan presented a biphasic expression, while Neurocan and Phosphacan expression declined dramatically with time and Versican expression remained broadly constant. In degeneration, Aggrecan expression increased markedly in Aipl1-/- and Pde6brd1/rd1, while Versican showed regional increases in the periphery of Rho-/- mice. Conversely, Neurocan and Phosphacan broadly decrease with time in all models. Our data reveal significant heterogeneity in the expression of individual CSPGs. Moreover, there are striking differences in the expression patterns of specific CSPGs in the diseased retina, compared with those reported following injury elsewhere in the CNS. Better understanding of the distinct distributions of individual CSPGs will contribute to creating more permissive microenvironments for neuro-regeneration and repair.


Subject(s)
Neurocan , Retinal Degeneration , Adaptor Proteins, Signal Transducing/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Animals , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Lectins, C-Type/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neurocan/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Versicans/genetics , Versicans/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...