Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Sci Rep ; 14(1): 1069, 2024 01 11.
Article in English | MEDLINE | ID: mdl-38212454

ABSTRACT

Salivary gland myoepithelial cells regulate saliva secretion and have been implicated in the histological diversity of salivary gland tumors. However, detailed functional analysis of myoepithelial cells has not been determined owing to the few of the specific marker to isolate them. We isolated myoepithelial cells from the submandibular glands of adult mice using the epithelial marker EpCAM and the cell adhesion molecule CD49f as indicators and found predominant expression of the transcription factor FoxO1 in these cells. RNA-sequence analysis revealed that the expression of cell cycle regulators was negatively regulated in FoxO1-overexpressing cells. Chromatin immunoprecipitation analysis showed that FoxO1 bound to the p21/p27 promoter DNA, indicating that FoxO1 suppresses cell proliferation through these factors. In addition, FoxO1 induced the expression of ectodysplasin A (Eda) and its receptor Eda2r, which are known to be associated with X-linked hypohidrotic ectodermal dysplasia and are involved in salivary gland development in myoepithelial cells. FoxO1 inhibitors suppressed Eda/Eda2r expression and salivary gland development in primordial organ cultures after mesenchymal removal. Although mesenchymal cells are considered a source of Eda, myoepithelial cells might be one of the resources of Eda. These results suggest that FoxO1 regulates myoepithelial cell proliferation and Eda secretion during salivary gland development in myoepithelial cells.


Subject(s)
Salivary Gland Neoplasms , Transcription Factors , Animals , Mice , Ectodysplasins/genetics , Epithelial Cells/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Salivary Gland Neoplasms/metabolism , Submandibular Gland/metabolism , Transcription Factors/metabolism , Xedar Receptor/metabolism
3.
Nature ; 617(7962): 827-834, 2023 May.
Article in English | MEDLINE | ID: mdl-37165186

ABSTRACT

Skeletal muscle atrophy is a hallmark of the cachexia syndrome that is associated with poor survival and reduced quality of life in patients with cancer1. Muscle atrophy involves excessive protein catabolism and loss of muscle mass and strength2. An effective therapy against muscle wasting is currently lacking because mechanisms driving the atrophy process remain incompletely understood. Our gene expression analysis in muscle tissues indicated upregulation of ectodysplasin A2 receptor (EDA2R) in tumour-bearing mice and patients with cachectic cancer. Here we show that activation of EDA2R signalling promotes skeletal muscle atrophy. Stimulation of primary myotubes with the EDA2R ligand EDA-A2 triggered pronounced cellular atrophy by induction of the expression of muscle atrophy-related genes Atrogin1 and MuRF1. EDA-A2-driven myotube atrophy involved activation of the non-canonical NFĸB pathway and was dependent on NFκB-inducing kinase (NIK) activity. Whereas EDA-A2 overexpression promoted muscle wasting in mice, deletion of either EDA2R or muscle NIK protected tumour-bearing mice from loss of muscle mass and function. Tumour-induced oncostatin M (OSM) upregulated muscle EDA2R expression, and muscle-specific oncostatin M receptor (OSMR)-knockout mice were resistant to tumour-induced muscle wasting. Our results demonstrate that EDA2R-NIK signalling mediates cancer-associated muscle atrophy in an OSM-OSMR-dependent manner. Thus, therapeutic targeting of these pathways may be beneficial in prevention of muscle loss.


Subject(s)
Cachexia , Muscular Atrophy , Neoplasms , Signal Transduction , Xedar Receptor , Animals , Mice , Cachexia/complications , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/pathology , Xedar Receptor/metabolism , Humans , Ligands , Receptors, Oncostatin M/metabolism , Oncostatin M/metabolism , NF-kappaB-Inducing Kinase
4.
Int J Radiat Biol ; 99(11): 1702-1715, 2023.
Article in English | MEDLINE | ID: mdl-37212632

ABSTRACT

PURPOSE: Previous research has highlighted the impact of radiation damage, with cancer patients developing acute disorders including radiation induced pneumonitis or chronic disorders including pulmonary fibrosis months after radiation therapy ends. We sought to discover biomarkers that predict these injuries and develop treatments that mitigate this damage and improve quality of life. MATERIALS AND METHODS: Six- to eight-week-old female C57BL/6 mice received 1, 2, 4, 8, 12 Gy or sham whole body irradiation. Animals were euthanized 48 h post exposure and lungs removed, snap frozen and underwent RNA isolation. Microarray analysis was performed to determine dysregulation of messenger RNA (mRNA), microRNA (miRNA), and long non-coding RNA (lncRNA) after radiation injury. RESULTS: We observed sustained dysregulation of specific RNA markers including: mRNAs, lncRNAs, and miRNAs across all doses. We also identified significantly upregulated genes that can indicate high dose exposure, including Cpt1c, Pdk4, Gdf15, and Eda2r, which are markers of senescence and fibrosis. Only three miRNAs were significantly dysregulated across all radiation doses: miRNA-142-3p and miRNA-142-5p were downregulated and miRNA-34a-5p was upregulated. IPA analysis predicted inhibition of several molecular pathways with increasing doses of radiation, including: T cell development, Quantity of leukocytes, Quantity of lymphocytes, and Cell viability. CONCLUSIONS: These RNA biomarkers might be highly relevant in the development of treatments and in predicting normal tissue injury in patients undergoing radiation treatment. We are conducting further experiments in our laboratory, which includes a human lung-on-a-chip model, to develop a decision tree model using RNA biomarkers.


Subject(s)
MicroRNAs , Whole-Body Irradiation , Mice , Animals , Humans , Whole-Body Irradiation/adverse effects , Quality of Life , Mice, Inbred C57BL , Lung/radiation effects , MicroRNAs/genetics , MicroRNAs/metabolism , Biomarkers/metabolism , Oligonucleotide Array Sequence Analysis , Disease Models, Animal , Xedar Receptor/genetics , Xedar Receptor/metabolism
5.
Int J Mol Sci ; 24(6)2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36982831

ABSTRACT

Animal models are invaluable in the research of the pathophysiology of interstitial cystitis/bladder pain syndrome (IC/BPS), a chronic aseptic urinary bladder disease of unknown etiology that primarily affects women. Here, a mouse model of IC/BPS was induced with multiple low-dose cyclophosphamide (CYP) applications and thoroughly characterized by RNA sequencing, qPCR, Western blot, and immunolabeling to elucidate key inflammatory processes and sex-dependent differences in the bladder inflammatory response. CYP treatment resulted in the upregulation of inflammatory transcripts such as Ccl8, Eda2r, and Vegfd, which are predominantly involved in innate immunity pathways, recapitulating the crucial findings in the bladder transcriptome of IC/BPS patients. The JAK/STAT signaling pathway was analyzed in detail, and the JAK3/STAT3 interaction was found to be most activated in cells of the bladder urothelium and lamina propria. Sex-based data analysis revealed that cell proliferation was more pronounced in male bladders, while innate immunity and tissue remodeling processes were the most distinctive responses of female bladders to CYP treatment. These processes were also reflected in prominent histological changes in the bladder. The study provides an invaluable reference dataset for preclinical research on IC/BPS and an insight into the sex-specific mechanisms involved in the development of IC/BPS pathology, which may explain the more frequent occurrence of this disease in women.


Subject(s)
Cystitis, Interstitial , Mice , Animals , Female , Male , Cystitis, Interstitial/genetics , Cystitis, Interstitial/pathology , Urinary Bladder/pathology , Transcriptome , Pelvis/pathology , Cell Proliferation , Disease Models, Animal , Xedar Receptor/metabolism
6.
Sci Rep ; 13(1): 200, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604457

ABSTRACT

Radiation injury from medical, accidental, or intentional sources can induce acute and long-term hepatic dysregulation, fibrosis, and cancer. This long-term hepatic dysregulation decreases quality of life and may lead to death. Our goal in this study is to determine acute changes in biological pathways and discover potential RNA biomarkers predictive of radiation injury. We performed whole transcriptome microarray analysis of mouse liver tissue (C57BL/6 J) 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray to identify significant expression changes in mRNAs, lncRNAs, and miRNAs, We also validated changes in specific RNAs through qRT-PCR. We used Ingenuity Pathway Analysis (IPA) to identify pathways associated with gene expression changes. We observed significant dysregulation of multiple mRNAs across all doses. In contrast, miRNA dysregulation was observed upwards of 2 Gray. The most significantly upregulated mRNAs function as tumor suppressors: Cdkn1a, Phlda3, and Eda2r. The most significantly downregulated mRNAs were involved in hemoglobin synthesis, inflammation, and mitochondrial function including multiple members of Hbb and Hba. The most significantly upregulated miRNA included: miR-34a-5p, miR-3102-5p, and miR-3960, while miR-342-3p, miR-142a-3p, and miR-223-3p were most significantly downregulated. IPA predicted activation of cell cycle checkpoint control pathways and inhibition of pathways relevant to inflammation and erythropoietin. Clarifying expression of mRNA, miRNA and lncRNA at a short time point (48 h) offers insight into potential biomarkers, including radiation markers shared across organs and animal models. This information, once validated in human models, can aid in development of bio-dosimetry biomarkers, and furthers our understanding of acute pathway dysregulation.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Mice , Inflammation , Liver/metabolism , Mice, Inbred C57BL , Microarray Analysis , MicroRNAs/genetics , MicroRNAs/metabolism , Quality of Life , RNA, Long Noncoding/genetics , Xedar Receptor
7.
Int J Biol Sci ; 18(15): 5624-5640, 2022.
Article in English | MEDLINE | ID: mdl-36263181

ABSTRACT

Diabetic kidney disease (DKD) is a major microvascular complication of diabetes mellitus and is one of the leading causes of end-stage kidney disease. Circular RNAs (circRNAs) are a class of endogenous non-coding RNAs that play important roles in various diseases, yet their roles in DKD are poorly understood. CircRNA HIPK3 (circHIPK3), a highly conserved circRNA, is closely related to various cellular functions, including cell proliferation and apoptosis. The association between circHIPK3 and diabetic complications has been well demonstrated in multiple previous studies. However, the role of circHIPK3 in podocyte injury in DKD remains unclear. Herein, we discovered that circHIPK3 expression is markedly elevated in cultured podocytes under high-glucose (HG) conditions and glomeruli of diabetic mice, which is closely associated with podocyte injury in DKD. Functionally, lentivirus-mediated knockdown of circHIPK3 dramatically suppresses HG-induced podocyte apoptosis in vitro. Therapeutically, silencing circHIPK3 by adeno-associated virus-mediated RNA interference ameliorates podocyte injury and albuminuria in STZ-induced diabetic mice. Mechanistically, circHIPK3 facilitates the enrichment of fused in sarcoma (FUS) on the ectodysplasin A2 receptor (EDA2R) promoter, resulting in the upregulation of EDA2R expression and activation of apoptotic signaling. Taken together, these results indicate circHIPK3/FUS/EDA2R axis as a therapeutic target for podocyte injury and DKD progression.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Podocytes , Mice , Animals , Podocytes/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , RNA, Circular/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Xedar Receptor/metabolism , Glucose/metabolism
8.
Allergol Immunopathol (Madr) ; 50(5): 84-90, 2022.
Article in English | MEDLINE | ID: mdl-36086968

ABSTRACT

BACKGROUND: Long-term hyperoxia impairs growth of the lungs and contributes to development of bronchopulmonary dysplasia. Ectodysplasin A (EDA) binds to ectodysplasin A2 receptor (EDA2R) and is essential for normal prenatal development. The functioning of EDA2R in bronchopulmonary dysplasia is investigated in this study. METHODS: Murine lung epithelial cells (MLE-12) were exposed to hyperoxia to induce cell injury. Cell viability and apoptosis were detected, respectively, by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay and flow cytometry. Inflammation and oxidative stress were evaluated by enzyme-linked immunosorbent serologic assay. RESULTS: Hyperoxia decreased cell viability and promoted cell apoptosis of MLE-12. EDA2R was elevated in hyperoxia-induced MLE-12. Silencing of EDA2R enhanced cell viability and reduced cell apoptosis of hyperoxia-induced MLE-12. Hyperoxia-induced up-regulation of tumor necrosis factor alpha (TNF-α), Interleukin (IL)-1ß, and IL-18 as well as MLE-12 was suppressed by knockdown of EDA2R. Inhibition of EDA2R down-regulated the level of malondialdehyde (MDA), up-regulated superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) in hyperoxia-induced MLE-12. Interference of EDA2R attenuated hyperoxia-induced increase in p-p65 in MLE-12. CONCLUSION: Knockdown of EDA2R exerted anti-inflammatory and antioxidant effects against hyperoxia-induced injury in lung epithelial cells through inhibition of nuclear factor kappa B (NF-κB) pathway.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Animals , Bronchopulmonary Dysplasia/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Infant, Newborn , Lung/pathology , Mice , NF-kappa B/metabolism , Xedar Receptor/metabolism
9.
Allergol. immunopatol ; 50(5): 84-90, sept. 2022. graf
Article in English | IBECS | ID: ibc-208629

ABSTRACT

Background: Long-term hyperoxia impairs growth of the lungs and contributes to develop-ment of bronchopulmonary dysplasia. Ectodysplasin A (EDA) binds to ectodysplasin A2 recep-tor (EDA2R) and is essential for normal prenatal development. The functioning of EDA2R in bronchopulmonary dysplasia is investigated in this study.Methods: Murine lung epithelial cells (MLE-12) were exposed to hyperoxia to induce cell injury. Cell viability and apoptosis were detected, respectively, by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay and flow cytometry. Inflammation and oxidative stress were evaluated by enzyme-linked immunosorbent serologic assay.Results: Hyperoxia decreased cell viability and promoted cell apoptosis of MLE-12. EDA2R was elevated in hyperoxia-induced MLE-12. Silencing of EDA2R enhanced cell viability and reduced cell apoptosis of hyperoxia-induced MLE-12. Hyperoxia-induced up-regulation of tumor necro-sis factor alpha (TNF-α), Interleukin (IL)-1β, and IL-18 as well as MLE-12 was suppressed by knockdown of EDA2R. Inhibition of EDA2R down-regulated the level of malondialdehyde (MDA), up-regulated superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) in hyperoxia-induced MLE-12. Interference of EDA2R attenuated hyperoxia-induced increase in p-p65 in M LE-12.Conclusion: Knockdown of EDA2R exerted anti-inflammatory and antioxidant effects against hyperoxia-induced injury in lung epithelial cells through inhibition of nuclear factor kappa B (NF-κB) pathway (AU)


Subject(s)
Animals , Bronchopulmonary Dysplasia/metabolism , Hyperoxia , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Lung/pathology , NF-kappa B/metabolism , Xedar Receptor/metabolism , Cell Line
10.
Am J Pathol ; 192(4): 613-628, 2022 04.
Article in English | MEDLINE | ID: mdl-35092726

ABSTRACT

Cisplatin induces both acute and chronic nephrotoxicity during chemotherapy in patients with cancer. Presented here is the first study of single-nucleus RNA sequencing (snRNA-seq) of cisplatin-induced nephrotoxicity. Repeated low-dose cisplatin treatment (RLDC) led to decreases in renal function and kidney weight in mice at 9 weeks. The kidneys of these mice showed tubular degeneration and dilation. snRNA-seq identified 16 cell types and 17 cell clusters in these kidneys. Cluster-by-cluster comparison demonstrated cell type-specific changes in gene expression and identified a unique proximal tubule (PT) injury/repair cluster that co-expressed the injury marker kidney injury molecule-1 (Kim1) and the proliferation marker Ki-67. Compared with control, post-RLDC kidneys had 424 differentially expressed genes in PT cells, including tubular transporters and cytochrome P450 enzymes involved in lipid metabolism. snRNA-seq also revealed transcriptional changes in potential PT injury markers (Krt222, Eda2r, Ltbp2, and Masp1) and repair marker (Bex4). RLDC induced inflammation and proinflammatory cytokines (RelB, TNF-α, Il7, Ccl2, and Cxcl2) and the expression of fibrosis markers (fibronectin, collagen I, connective tissue growth factor, vimentin, and α-smooth muscle actin). Together, these results provide new insights into RLDC-induced transcriptional changes at the single-cell level that may contribute to the development of chronic kidney problems in patients with cancer after cisplatin chemotherapy.


Subject(s)
Acute Kidney Injury , Antineoplastic Agents , Renal Insufficiency, Chronic , Acute Kidney Injury/pathology , Animals , Biomarkers/metabolism , Cisplatin/toxicity , Fibrosis , Humans , Kidney/pathology , Latent TGF-beta Binding Proteins/metabolism , Mice , RNA, Small Nuclear/metabolism , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Xedar Receptor/metabolism
11.
Commun Biol ; 4(1): 213, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594251

ABSTRACT

The mucosa microenvironment is critical for intestinal stem cell self-renewal and reconstruction of the epithelial barrier in inflammatory bowel disease (IBD), where the mechanisms underlying cross-talk between intestinal crypts and the microenvironment remain unclear. Here, we firstly identified miR-494-3p as an important protector in colitis. miR-494-3p levels were decreased and negatively correlated with the severity in human IBD samples, as well as in colitis mice. In colitis crypts, a notable cytokine-cytokine receptor, miR-494-3p-targeted EDA2R and the ligand EDA-A2, suppressed colonic stemness and epithelial repair by inhibiting ß-catenin/c-Myc. In differentiated IECs, miR-494-3p inhibits macrophage recruitment, M1 activation and EDA-A2 secretion by targeting IKKß/NF-κB in colitis. A miR-494-3p agomir system notably ameliorated the severity of colonic colitis in vivo. Collectively, our findings uncover a miR-494-3p-mediated cross-talk mechanism by which macrophage-induced intestinal stem cell impairment aggravates intestinal inflammation.


Subject(s)
Colitis/metabolism , Colon/metabolism , Ectodysplasins/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Paracrine Communication , Stem Cells/metabolism , Xedar Receptor/metabolism , Animals , Antagomirs/administration & dosage , Cells, Cultured , Chemotaxis , Colitis/genetics , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Disease Models, Animal , Ectodysplasins/genetics , Humans , I-kappa B Kinase/metabolism , Intestinal Mucosa/pathology , Macrophage Activation , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Organoids , Stem Cell Niche , Stem Cells/pathology , Wnt Signaling Pathway , Xedar Receptor/genetics
12.
Cell Transplant ; 30: 963689721996346, 2021.
Article in English | MEDLINE | ID: mdl-33637015

ABSTRACT

X-linked ectodermal dysplasia receptor (XEDAR) is a new member of the tumor necrosis factor receptor (TNFR) family that induces cell death. The purpose of this study is to determine the tumor-suppressive potential of XEDAR in the development and differentiation of gastric cancer (GC). XEDAR levels were analyzed in human GC tissues and adjacent normal tissues by immunohistochemistry (IHC), quantitative real-time reverse transcription PCR (RT-qPCR), and Western blot analysis. We found that XEDAR expression was significantly downregulated in GC tissues and further decreased in low differentiated GC tissues. Overexpression of XEDAR in MKN45 and MGC803 cells suppressed the ability of cell proliferation and migration, whereas silencing XEDAR showed the opposite effect. Additionally, XEDAR silencing resulted in the upregulation of the differentiation molecular markers ß-catenin, CD44 and Cyclin D1 at the protein levels, whereas XEDAR overexpression showed the opposite effect. Notably, XEDAR positively regulated the expression of liver X receptor alpha (LXRα) through upregulating the RELA gene that was characterized as a transcription factor of LXRα in this study. Inhibition of LXRα by GSK2033 or activation of the Wnt/ß-catenin pathway by Wnt agonist 1 impaired the effect of XEDAR overexpression on differentiation of MKN45 cells. Moreover, inhibition of RELA mediated by siRNA could promote cell proliferation/migration and rescue the effect of XEDAR overexpression on cell behaviors and expression of genes. Subsequently, overexpression of XEDAR suppressed the growth of GC cells in vivo. Taken together, our findings showed that XEDAR could promote differentiation and suppress proliferation and invasion of GC cells.


Subject(s)
Liver X Receptors/metabolism , Stomach Neoplasms/metabolism , Transcription Factor RelA/metabolism , Wnt Signaling Pathway , Xedar Receptor/metabolism , beta Catenin/metabolism , Adult , Aged , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Female , Genes, Tumor Suppressor , Humans , Liver X Receptors/genetics , Male , Middle Aged , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factor RelA/genetics , Up-Regulation , Xedar Receptor/genetics , beta Catenin/genetics
13.
Biochem Biophys Res Commun ; 529(3): 766-772, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32736705

ABSTRACT

Androgenetic alopecia (AGA) is a common genetic disorder, and a X-chromosomal locus that contains the androgen receptor (AR) and ectodysplasin A2 receptor (EDA2R) genes represents a major susceptibility locus for AGA. In our previous study, we reported that ectodysplasin-A2 (EDA-A2) induces apoptosis in cultured human hair follicle (HF) cells and promotes the regression of HFs in mice. However, the role of the EDA-A2/EDA2R in AGA remains unknown, as the causative gene in this pathway has not yet been identified and potential functional connections between EDA-A2 signaling and the androgen pathway remain unclear. In this study, we investigated the expression of EDA2R in balding HFs and matched with non-balding HFs. The EDA2R level was upregulated in the balding dermal papilla (DP) cells compared with non-balding DP cells derived from patients with AGA. However, EDA2R was strongly expressed in both balding and non-balding outer root sheath (ORS) cells. We screened EDA-A2-regulated genes in balding DP cells and identified dickkopf 1 (DKK-1) as catagen inducer during the hair cycle. The mRNA and protein expression levels of DKK-1 were both upregulated by EDA-A2. In addition, DKK-1 expression was induced by EDA-A2 both in cultured human HFs and in mouse HFs. Moreover, the EDA-A2-induced apoptosis of DP and ORS cells was reversed by the antibody-mediated neutralization of DKK-1. Collectively, our data strongly suggest that EDA-A2 induces DKK-1 secretion and causes apoptosis in HFs by binding EDA2R, which is overexpressed in the bald scalp. EDA-A2/EDA2R signaling could inhibit hair growth through DKK-1 induction, and an inhibitor of EDA-A2/EDA2R signaling may be a promising agent for the treatment and prevention of AGA.


Subject(s)
Alopecia/genetics , Ectodysplasins/metabolism , Hair Follicle/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Xedar Receptor/metabolism , Alopecia/metabolism , Apoptosis , Cells, Cultured , Hair Follicle/cytology , Humans , Up-Regulation , Xedar Receptor/genetics
14.
Biochimie ; 174: 74-83, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32304771

ABSTRACT

EDA2R is a member of the large family of tumor necrosis factor receptor (TNFR). Previous studies suggested that EDA2R expression might be increased in the kidneys of diabetic mice. However, its mRNA and protein expression in kidneys were not analyzed; moreover, its role in the development of diabetic kidney disease was not explored. Here we analyzed the mRNA and protein expressions of EDA2R in diabetic kidneys and examined its role in the podocyte injury in high glucose milieu. By analysis with real-time PCR, Western blotting, we found that both the mRNA and protein levels of EDA2R were increased in the kidneys of diabetic mice. Immunohistochemical studies revealed that EDA2R expression was enhanced in both glomerular and tubular cells of diabetic mice and humans. In vitro studies, high glucose increased EDA2R expression in cultured human podocytes. Overexpression of EDA2R in podocytes promoted podocyte apoptosis and decreased nephrin expression. Moreover, ED2AR increased ROS generation in podocytes, while inhibiting ROS generation attenuates EDA2R-mediated podocyte injury. In addition, EDA2R silencing partially suppressed high glucose-induced ROS generation, apoptosis, and nephrin decrease. Our study demonstrated that high glucose increases EDA2R expression in kidney cells and that EDA2R induces podocyte apoptosis and dedifferentiation in high glucose milieu partially through enhanced ROS generation.


Subject(s)
Diabetes Mellitus/metabolism , Diabetic Nephropathies/metabolism , Kidney/metabolism , Podocytes/metabolism , Xedar Receptor/physiology , Animals , Apoptosis , Cells, Cultured , Female , Kidney/pathology , Membrane Proteins/metabolism , Mice , Podocytes/pathology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
15.
Oxid Med Cell Longev ; 2019: 3829342, 2019.
Article in English | MEDLINE | ID: mdl-31885785

ABSTRACT

Little information has been available about the influence of dietary genistein (GEN) on hepatic transcriptome of laying broiler breeder (LBB) hens. The study is aimed at broadening the understanding of RNA expression profiles and alternative splicing (AS) signatures of GEN-treated breeder hens and thereby improving laying performance and immune function of hens during the late egg-laying period. 720 LBB hens were randomly allocated into three groups with supplemental dietary GEN doses (0, 40 mg/kg, and 400 mg/kg). Each treatment has 8 replicates of 30 birds. Dietary GEN enhanced the antioxidative capability of livers, along with the increased activities of glutathione peroxidase and catalase. Furthermore, it improved lipid metabolic status and apoptotic process in the liver of hens. 40 mg/kg dietary GEN had the better effects on improving immune function and laying performance. However, transcriptome data indicated that 400 mg/kg dietary GEN did negative regulation of hormone biosynthetic process. Also, it upregulated the expressions of EDA2R and CYR61 by the Cis regulation of neighbouring genes (lncRNA_XLOC_018890 and XLOC_024242), which might activate NF-κB and immune-related signaling pathway. Furthermore, dietary GEN induced AS events in the liver, which also enriched into immune and metabolic process. Therefore, the application of 40 mg/kg GEN in the diet of breeder hens during the late egg-laying period can improve lipid metabolism and immune function. We need to pay attention to the side-effects of high-dose GEN on the immune function.


Subject(s)
Alternative Splicing/drug effects , Genistein/pharmacology , Liver/drug effects , RNA/metabolism , Transcriptome/drug effects , Animals , Antioxidants/metabolism , Chickens , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Dietary Supplements , Liver/metabolism , RNA, Long Noncoding/metabolism , Triglycerides/blood , Xedar Receptor/genetics , Xedar Receptor/metabolism
16.
Biosci Rep ; 39(12)2019 12 20.
Article in English | MEDLINE | ID: mdl-31829409

ABSTRACT

X-linked ectodermal dysplasia receptor (XEDAR) has been widely studied in epidermal morphogenesis, but few studies have been conducted on tumorigenesis and development, including gastric cancer. In the present research, we aimed to investigate the effect of XEDAR on gastric cancer and further explore the molecular mechanisms involved. The differential expression of XEDAR in 90 tissue specimens (30 gastric cancer tissues, 30 adjacent tissues and 30 normal tissues) was detected by real-time PCR (RT-PCR) and Western blot. Cell proliferation and apoptosis were explored using MTT and Annexin-V/propidium iodide (PI) assays, respectively. The results revealed that the expression of XEDAR was decreased in gastric cancer tissues and in gastric cancer cell lines, and its expression is regulated by p53 in BGC-823 cells. Furthermore, overexpression of XEDAR inhibited cell proliferation and induced apoptosis in BGC-823 cells. XEDAR moreover inhibited proliferation and induced apoptosis in gastric cancer cells by regulating the JNK signaling pathway. Collectively, the results of the present study suggested that XEDAR inhibits cell proliferation and induces apoptosis by participating in p53-mediated signaling pathway and inhibiting the downstream JNK signaling pathway in gastric cancer.


Subject(s)
Apoptosis , Cell Proliferation , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Xedar Receptor/metabolism , Cell Line, Tumor , Humans , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Xedar Receptor/genetics
17.
Biochem Biophys Res Commun ; 520(2): 428-433, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31607478

ABSTRACT

Ectodysplasin is a ligand of the TNF family that plays a key role in ectodermal differentiation. EDA-A1 and EDA-A2 are two isoforms of ectodysplasin that differ only by the insertion of two amino acids and bind to two different receptors, ectodysplasin A receptor (EDAR) and ectodysplasin A2 receptor (EDA2R), respectively. Mutations of EDA-A1 and its receptor EDAR have been associated with hypohidrotic ecodermal dysplasia (HED). However, the role of EDA-A2 and the expression pattern of EDA2R in human hair follicles and in the mouse hair growth cycle have not been reported. In this study, we first investigated the expression of EDA2R in human hair follicles and in cultured follicular cells. EDA2R was strongly expressed in outer root sheath (ORS) cells and weakly expressed in dermal papilla (DP) cells. EDA-A2 induced the apoptosis of both ORS cells and DP cells via the activation of cleaved caspase-3. In addition, EDA2R was highly expressed in the late anagen phase compared with other phases in the hair growth cycle. Moreover, EDA-A2 induced apoptosis in cultured human hair follicle cells and in the mouse hair growth cycle, causing the premature onset of the catagen phase. Collectively, our results suggest that EDA-A2/EDA2R signaling could inhibit hair growth, and an inhibitor of EDA-A2/EDA2R signaling may be a promising agent for the treatment and prevention of hair loss.


Subject(s)
Ectodysplasins/pharmacology , Hair Follicle/cytology , Xedar Receptor/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Cells, Cultured , Ectodysplasins/genetics , Ectodysplasins/metabolism , Female , Hair Follicle/drug effects , Humans , Male , Mice, Inbred C57BL , Xedar Receptor/genetics
18.
Eur J Prev Cardiol ; 25(1_suppl): 51-58, 2018 06.
Article in English | MEDLINE | ID: mdl-29708036

ABSTRACT

Background Sporadic non-syndromic thoracic aortic aneurysms (SNSTAAs) are less well understood than familial non-syndromic or syndromic ones. Here, we focused on morphologic and molecular changes of the extracellular matrix of the tunica media of SNSTAAs. Design Single centre design. Methods Surgical media samples from seven SNSTAAs and seven controls underwent quantitative polymerase chain reaction, proteomics-bioinformatics, immunoblotting, histology and immunohistochemistry analysis. Results A down-regulation of Decorin mRNA with unchanged protein levels associated with a remarkable increase of collagen fibres. A reduced and distorted network of elastic fibres partnered with an attenuated expression of microfibril-associated glycoprotein1 despite the rise of MFAP2 gene-encoded mRNA levels. An increasingly proteolysed paxillin (55 kDa PXN), a focal adhesion protein, combined with an upregulated 62 kDa PXN holoprotein, without changes in amount and phosphorylation of focal adhesion kinase (pp125FAK). The upregulation of SPOCK2-encoded Testican2 proteoglycan and of ectodysplasin (EDA) protein was coupled with a down-regulation of EDA2 receptor (EDA2R). Conclusions Several tunica media extracellular matrix-related changes favour SNSTAA development. A steady level of decorin and a microfibril-associated glycoprotein1 protein shortage cause the assembly of structurally defective collagen and elastic fibres. Up-regulation of PXN holoproteins perturbs PXN/pp125FAK interaction and focal adhesion functioning. Testican2 up-regulation suppresses the membrane-type matrix metalloproteinase inhibiting activities of other SPOCK family members thus enhancing extracellular matrix proteolysis. Finally, the altered EDA•EDA2R signalling would impact on the remodelling of SNSTAA tunica media. Altogether, our results pave the way to a deeper molecular understanding of SNSTAAs necessary to identify their early diagnostic biochemical markers.


Subject(s)
Aortic Aneurysm, Thoracic/genetics , Decorin/genetics , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Gene Expression Regulation , Proteoglycans/genetics , Xedar Receptor/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Decorin/biosynthesis , Extracellular Matrix/pathology , Humans , Immunoblotting , Immunohistochemistry , Polymerase Chain Reaction , Proteoglycans/biosynthesis , RNA/genetics , Xedar Receptor/biosynthesis
19.
Clin Exp Med ; 17(1): 111-119, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26659383

ABSTRACT

Despite recent advancements in the knowledge of the etiology and pathogenic mechanisms, treatment of the autoimmune disease Sjögren's syndrome (SS) remains mostly empiric and symptom-based, indicating the need for novel therapeutic approaches. Ectodysplasin-A2 (EDA-A2) is a recently isolated member of the tumor necrosis factor superfamily that binds to X-linked ectodermal dysplasia receptor (XEDAR). In this report, we have analyzed the expression and the biological activity of EDA-A2 in human salivary gland epithelial cells (SGEC) from primary Sjögren's syndrome (pSS) patients. We report that EDA-A2 and its receptor XEDAR are overexpressed in pSS SGEC in comparison with healthy individuals and that the EDA-A2/XEDAR system in these cells is involved in the induction of apoptosis via caspases activation. Collectively, our results suggest that EDA-A2/XEDAR system may be a promising agent for the gene therapy of pSS.


Subject(s)
Caspase 3/genetics , Ectodysplasins/genetics , Epithelial Cells/metabolism , Salivary Glands/metabolism , Sjogren's Syndrome/genetics , Xedar Receptor/genetics , Apoptosis/genetics , Case-Control Studies , Caspase 3/metabolism , Ectodysplasins/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Salivary Glands/pathology , Signal Transduction , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology , Xedar Receptor/antagonists & inhibitors , Xedar Receptor/metabolism
20.
J Invest Dermatol ; 136(8): 1559-1567, 2016 08.
Article in English | MEDLINE | ID: mdl-27060448

ABSTRACT

Androgenetic alopecia (AGA) is a common heritable and androgen-dependent hair loss condition in men. Twelve genetic risk loci are known to date, but it is unclear which genes at these loci are relevant for AGA. Dermal papilla cells (DPCs) located in the hair bulb are the main site of androgen activity in the hair follicle. Widely used monolayer-cultured primary DPCs in hair-related studies often lack dermal papilla characteristics. In contrast, immortalized DPCs have high resemblance to intact dermal papilla. We derived immortalized human DPC lines from balding (BAB) and non-balding (BAN) scalp. Both BAB and BAN retained high proportions of dermal papilla signature gene and versican protein expression. We performed expression analysis of BAB and BAN and annotated AGA risk loci with differentially expressed genes. We found evidence for AR but not EDA2R as the candidate gene at the AGA risk locus on chromosome X. Further, our data suggest TWIST1 (twist family basic helix-loop-helix transcription factor 1) and SSPN (sarcospan) to be the functionally relevant AGA genes at the 7p21.1 and 12p12.1 risk loci, respectively. Down-regulated genes in BAB compared to BAN were highly enriched for vasculature-related genes, suggesting that deficiency of DPC from balding scalps in fostering vascularization around the hair follicle may contribute to the development of AGA.


Subject(s)
Alopecia/genetics , Dermis/cytology , Gene Expression Regulation , Skin/cytology , Androgens/metabolism , Biopsy , Carrier Proteins/genetics , Cell Line , Cell Nucleus/metabolism , Cluster Analysis , Gene Expression Profiling , Hair Follicle/metabolism , Humans , Male , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Receptors, Androgen/genetics , Scalp , Twist-Related Protein 1/genetics , Xedar Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...