Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 617(7962): 827-834, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37165186

RESUMEN

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.


Asunto(s)
Caquexia , Atrofia Muscular , Neoplasias , Transducción de Señal , Receptor Xedar , Animales , Ratones , Caquexia/complicaciones , Caquexia/etiología , Caquexia/metabolismo , Caquexia/patología , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Atrofia Muscular/prevención & control , Neoplasias/complicaciones , Neoplasias/metabolismo , Neoplasias/patología , Receptor Xedar/metabolismo , Humanos , Ligandos , Receptores de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Quinasa de Factor Nuclear kappa B
2.
Am J Pathol ; 192(4): 613-628, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35092726

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda , Antineoplásicos , Insuficiencia Renal Crónica , Lesión Renal Aguda/patología , Animales , Biomarcadores/metabolismo , Cisplatino/toxicidad , Fibrosis , Humanos , Riñón/patología , Proteínas de Unión a TGF-beta Latente/metabolismo , Ratones , ARN Nuclear Pequeño/metabolismo , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Receptor Xedar/metabolismo
3.
Int J Mol Sci ; 24(6)2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36982831

RESUMEN

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.


Asunto(s)
Cistitis Intersticial , Ratones , Animales , Femenino , Masculino , Cistitis Intersticial/genética , Cistitis Intersticial/patología , Vejiga Urinaria/patología , Transcriptoma , Pelvis/patología , Proliferación Celular , Modelos Animales de Enfermedad , Receptor Xedar/metabolismo
4.
Allergol Immunopathol (Madr) ; 50(5): 84-90, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36086968

RESUMEN

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.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Animales , Displasia Broncopulmonar/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Hiperoxia/patología , Recién Nacido , Pulmón/patología , Ratones , FN-kappa B/metabolismo , Receptor Xedar/metabolismo
5.
Biochem Biophys Res Commun ; 529(3): 766-772, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32736705

RESUMEN

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.


Asunto(s)
Alopecia/genética , Ectodisplasinas/metabolismo , Folículo Piloso/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Receptor Xedar/metabolismo , Alopecia/metabolismo , Apoptosis , Células Cultivadas , Folículo Piloso/citología , Humanos , Regulación hacia Arriba , Receptor Xedar/genética
6.
Biochem Biophys Res Commun ; 520(2): 428-433, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31607478

RESUMEN

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.


Asunto(s)
Ectodisplasinas/farmacología , Folículo Piloso/citología , Receptor Xedar/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3/metabolismo , Células Cultivadas , Ectodisplasinas/genética , Ectodisplasinas/metabolismo , Femenino , Folículo Piloso/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Receptor Xedar/genética
7.
Biochem Biophys Res Commun ; 465(2): 275-80, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26260321

RESUMEN

Members of the tumor necrosis factor receptor (TNFR) superfamily are involved in a number of physiological and pathological responses by activating a wide variety of intracellular signaling pathways. The X-linked ectodermal dysplasia receptor (XEDAR; also known as EDA2R or TNFRSF27) is a member of the TNFR superfamily that is highly expressed in ectodermal derivatives during embryonic development and binds to ectodysplasin-A2 (EDA-A2), a member of the TNF family that is encoded by the anhidrotic ectodermal dysplasia (EDA) gene. Although XEDAR was first described in the year 2000, its function and molecular mechanism of action is still largely unclear. XEDAR has been reported to activate canonical nuclear factor κB (NF-κB) signaling and mitogen-activated protein (MAP) kinases. Here we report that XEDAR is also able to trigger the non-canonical NF-κB pathway, characterized by the processing of p100 (NF-κB2) into p52, followed by nuclear translocation of p52 and RelB. We provide evidence that XEDAR-induced p100 processing relies on the binding of XEDAR to TRAF3 and TRAF6, and requires the kinase activity of NIK and IKKα. We also show that XEDAR stimulation results in NIK accumulation and that p100 processing is negatively regulated by TRAF3, cIAP1 and A20.


Asunto(s)
Regulación de la Expresión Génica , Subunidad p52 de NF-kappa B/genética , Receptor Xedar/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ectodisplasinas/genética , Ectodisplasinas/metabolismo , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Receptor Xedar/metabolismo , Quinasa de Factor Nuclear kappa B
8.
J Neurogenet ; 29(4): 188-94, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26814133

RESUMEN

Migraine without aura (MWO) is the most common among migraine group, and is mainly associated with genetic, physical and chemical factors, and hormonal changes. We aimed to identify novel non-synonymous mutations predisposing to the susceptibility to MWO in a Chinese sample using exome sequencing. Four patients with MWO from a family and four non-migraine subjects unrelated with these patients were genotyped using whole-exome sequencing. Bioinformatics analysis was used to screen possible susceptibility gene mutations, which were then verified by PCR. In four patients with MWO, six novel rare non-synonymous mutations were observed, including EDA2R (G170A), UBE2NL (T266G), GBP2 (A907G), EMR1 (C264G), CLCNKB (A1225G), and ARHGAP28 (C413G). It is worth stressing that GBP2 (A907G) was absent in any control subject. Multiple genes predispose to the susceptibility to MWO. ARHGAP28-, EMR1-, and GBP2-encoded proteins may affect angiokinesis, which supports vasogenic theory for the etiological hypothesis of this disease. CLCNKB-encoded protein may affect cell membrane potential, which is consistent with the cortical spreading depression theory. UBE2NL-encoded protein may regulate cellular responses to 5-hydroxytryptamine, which is in accordance with trigeminovascular reflex theory. EDA2R and UBE2NL are located on the X chromosome, which supports that this disease may have gender differences in genetic predisposition. Replication in larger sample size would significantly strengthen these findings.


Asunto(s)
Predisposición Genética a la Enfermedad , Migraña sin Aura/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al Calcio , Canales de Cloruro/genética , Biología Computacional , Análisis Mutacional de ADN , Exoma/genética , Salud de la Familia , Femenino , Proteínas de Unión al GTP/genética , Proteínas Activadoras de GTPasa/genética , Estudios de Asociación Genética , Humanos , Masculino , Glicoproteínas de Membrana/genética , Mucinas/genética , Receptores Acoplados a Proteínas G/genética , Enzimas Ubiquitina-Conjugadoras/genética , Receptor Xedar/genética
9.
Sci Rep ; 14(1): 1069, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212454

RESUMEN

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.


Asunto(s)
Neoplasias de las Glándulas Salivales , Factores de Transcripción , Animales , Ratones , Ectodisplasinas/genética , Células Epiteliales/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Neoplasias de las Glándulas Salivales/metabolismo , Glándula Submandibular/metabolismo , Factores de Transcripción/metabolismo , Receptor Xedar/metabolismo
10.
Gynecol Oncol ; 130(3): 565-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23800698

RESUMEN

OBJECTIVE: The objective of this study is to investigate whether wild-type TP53 status in high-grade serous ovarian carcinoma is associated with poorer survival. METHODS: Clinical and genomic data of 316 sequenced samples from The Cancer Genome Atlas (TCGA) ovarian high-grade serous carcinoma study were downloaded from TCGA data portal. Association between wild-type TP53 and survival was analyzed with Kaplan Meier method and Cox regression. The diagnosis of high-grade serous carcinomas was evaluated by reviewing pathological reports and high-resolution hematoxylin and eosin (H&E) images from frozen sections. The authenticity of wild-type TP53 in these tumor samples was assessed by analyzing SNP array data with ASCAT algorithm, reverse phase protein array (RPPA) data and RNAseq data. RESULTS: Fifteen patients with high grade serous ovarian carcinomas were identified to have wild-type TP53, which had significantly shorter survival and higher chemoresistance than those with mutated TP53. The authenticity of wild-type TP53 status in these fifteen patients was supported by SNP array, RPPA, and RNAseq data. Except four cases with mixed histology, the classification as high grade serous carcinomas was supported by pathological reports and H&E images. Using RNAseq data, it was found that EDA2R gene, a direct target of wild-type TP53, was highly up-regulated in samples with wild-type TP53 in comparison to samples with either nonsense or missense TP53 mutations. CONCLUSION: Although patients with wild-type TP53 ovarian cancer were rare in the TCGA high grade ovarian serous carcinomas cohort, these patients appeared to have a poorer survival and were more chemoresistant than those with mutated TP53. Differentially expressed genes in these TP53 wild-type tumors may provide insight in the molecular mechanism in chemotherapy resistance.


Asunto(s)
Cistadenocarcinoma Seroso/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes p53/genética , Neoplasias Ováricas/genética , Receptor Xedar/genética , Cistadenocarcinoma Seroso/patología , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Ováricas/patología , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Análisis por Matrices de Proteínas , Análisis de Secuencia de ARN , Regulación hacia Arriba
11.
Int J Radiat Biol ; 99(11): 1702-1715, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37212632

RESUMEN

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.


Asunto(s)
MicroARNs , Irradiación Corporal Total , Ratones , Animales , Humanos , Irradiación Corporal Total/efectos adversos , Calidad de Vida , Ratones Endogámicos C57BL , Pulmón/efectos de la radiación , MicroARNs/genética , MicroARNs/metabolismo , Biomarcadores/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Modelos Animales de Enfermedad , Receptor Xedar/genética , Receptor Xedar/metabolismo
12.
Sci Rep ; 13(1): 200, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36604457

RESUMEN

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.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Animales , Ratones , Inflamación , Hígado/metabolismo , Ratones Endogámicos C57BL , Análisis por Micromatrices , MicroARNs/genética , MicroARNs/metabolismo , Calidad de Vida , ARN Largo no Codificante/genética , Receptor Xedar
13.
Br J Dermatol ; 166(6): 1314-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22309448

RESUMEN

BACKGROUND: The aetiology of female pattern hair loss (FPHL) is largely unknown. However, it is hypothesized that FPHL and male pattern baldness (AGA) share common susceptibility alleles. The two major susceptibility loci for AGA are the androgen receptor (AR)/ectodysplasin A2 receptor (EDA2R) locus on the X-chromosome, and a locus on chromosome 20p11, for which no candidate gene has yet been identified. OBJECTIVES: To examine the role of the AR/EDA2R and 20p11 loci in the development of FPHL using 145 U.K. and 85 German patients with FPHL, 179 U.K. supercontrols and 150 German blood donors. METHODS: Patients and controls were genotyped for 25 single nucleotide polymorphisms (SNPs) at the AR/EDA2R locus and five SNPs at the 20p11 locus. RESULTS: Analysis of the AR/EDA2R locus revealed no significant association in the German sample. However, a nominally significant association for a single SNP (rs1397631) was found in the U.K. sample. Subgroup analysis of the U.K. patients revealed significant association for seven markers in patients with an early onset (P = 0·047 after adjustment for the testing of multiple SNPs by Monte Carlo simulation). No significant association was obtained for the five 20p11 variants, either in the overall samples or in the analysis of subgroups. CONCLUSIONS: The observed association suggests that the AR/EDA2R locus confers susceptibility to early-onset FHPL. Our results do not implicate the 20p11 locus in the aetiology of FPHL.


Asunto(s)
Alopecia/genética , Cromosomas Humanos Par 20/genética , Polimorfismo de Nucleótido Simple/genética , Receptor Xedar/genética , Estudios de Casos y Controles , Femenino , Sitios Genéticos , Predisposición Genética a la Enfermedad/genética , Humanos , Persona de Mediana Edad
14.
Hum Biol ; 84(6): 641-94, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23959643

RESUMEN

Single nucleotide polymorphisms (SNPs) with large allele frequency differences between human populations are relatively rare. The longest run of SNPs with an allele frequency difference of one between the Yoruba of Nigeria and the Han Chinese is found on the long arm of the X chromosome in the intergenic region separating the EDA2R and AR genes. It has been proposed that the unusual allele frequency distributions of these SNPs are the result of a selective sweep affecting African populations that occurred after the out-of-Africa migration. To investigate the evolutionary history of the EDA2R/AR intergenic region, we characterized the haplotype structure of 52 of its highly differentiated SNPs. Using a publicly available data set of 3,000 X chromosomes from 65 human populations, we found that nearly all human X chromosomes carry one of two modal haplotypes for these 52 SNPs. The predominance of two highly divergent haplotypes at this locus was confirmed by use of a subset of individuals sequenced to high coverage. The first of these haplotypes, the α-haplotype is at high frequencies in most of the African populations surveyed and likely arose before the separation of African populations into distinct genetic entities. The second, the ß-haplotype, is frequent or fixed in all non-African populations and likely arose in East Africa before the out-of-Africa migration. We also observed a small group or rare haplotypes with no clear relationship to the α- and ß-haplotypes. These haplotypes occur at relatively high frequencies in African hunter-gatherer populations, such as the San and Mbuti Pygmies. Our analysis indicates that these haplotypes are part of a pool of diverse, ancestral haplotypes that have now been almost entirely replaced by the α- and ß-haplotypes. We suggest that the rise of the α- and ß-haplotypes was the result of the demographic forces that human populations experienced during the formation of modern African populations and the out-of-Africa migration. However, we also present evidence that this region is the target of selection in the form of positive selection on the α- and ß-haplotypes and of purifying the selection against α/ß recombinants.


Asunto(s)
Pueblo Asiatico/genética , Población Negra/genética , ADN Intergénico/genética , Genoma Humano , Haplotipos/genética , Receptores Androgénicos/genética , Receptor Xedar/genética , Alelos , Evolución Biológica , Cromosomas Humanos X , Femenino , Frecuencia de los Genes , Genética de Población , Humanos , Masculino , Polimorfismo de Nucleótido Simple
15.
Int J Biol Sci ; 18(15): 5624-5640, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36263181

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Podocitos , Ratones , Animales , Podocitos/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , ARN Circular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Receptor Xedar/metabolismo , Glucosa/metabolismo
16.
Am J Orthod Dentofacial Orthop ; 140(1): e9-15, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21724072

RESUMEN

INTRODUCTION: Dental crowding is a problem for both adolescents and adults in modern society. The purpose of this research was to identify single nucleotide polymorphisms (SNPs) responsible for crowding in subjects with skeletal Class I relationships. METHODS: The case subjects consisted of healthy Chinese people living in Hong Kong with skeletal Class I relationships and at least 5 mm of crowding in either arch. The control subjects met the same requirements but lacked crowding or spacing. SNP genotyping was performed on the MassARRAY platform. The chi-square test was used to compare genotype and allele type distributions between the case and the control groups. Logistic regression was used to calculate odds ratios with 95% confidence intervals, and the effects of age and sex for each SNP. Analyses of linkage disequilibrium and haplotype associations between SNPs were performed with software. RESULTS: Five SNPs were found to be significantly different in genotype or allele type distributions. SNP rs372024 was significantly associated with crowding (P = 0.004). Two SNPs, rs3764746 and rs3795170, on the EDA gene were found to be associated marginally. SNPs rs1005464 and rs15705 also exhibited marginal association with crowding. The effects of associated SNPs remained significant after adjustments for age and sex factors. CONCLUSIONS: This study suggests an association for the genes EDA and XEDAR in dental crowding in the Hong Kong Chinese population.


Asunto(s)
Ectodisplasinas/genética , Maloclusión Clase I de Angle/genética , Polimorfismo de Nucleótido Simple , Receptor Xedar/genética , Adolescente , Pueblo Asiatico/genética , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Análisis Mutacional de ADN/métodos , Femenino , Frecuencia de los Genes , Hong Kong , Humanos , Desequilibrio de Ligamiento , Modelos Logísticos , Masculino , Espectrometría de Masas/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
17.
Cell Transplant ; 30: 963689721996346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33637015

RESUMEN

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.


Asunto(s)
Receptores X del Hígado/metabolismo , Neoplasias Gástricas/metabolismo , Factor de Transcripción ReIA/metabolismo , Vía de Señalización Wnt , Receptor Xedar/metabolismo , beta Catenina/metabolismo , Adulto , Anciano , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Genes Supresores de Tumor , Humanos , Receptores X del Hígado/genética , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factor de Transcripción ReIA/genética , Regulación hacia Arriba , Receptor Xedar/genética , beta Catenina/genética
18.
Commun Biol ; 4(1): 213, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594251

RESUMEN

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.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Ectodisplasinas/metabolismo , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Comunicación Paracrina , Células Madre/metabolismo , Receptor Xedar/metabolismo , Animales , Antagomirs/administración & dosificación , Células Cultivadas , Quimiotaxis , Colitis/genética , Colitis/patología , Colitis/prevención & control , Colon/patología , Modelos Animales de Enfermedad , Ectodisplasinas/genética , Humanos , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/patología , Activación de Macrófagos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Organoides , Nicho de Células Madre , Células Madre/patología , Vía de Señalización Wnt , Receptor Xedar/genética
19.
Exp Dermatol ; 19(11): 1026-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21073542

RESUMEN

The gene encoding the androgen receptor (AR) is associated with male pattern baldness (androgenetic alopecia - AGA). In case-control and family analyses, we mapped AR and the adjacent intergenic regions. We found evidence for association with two independent loci, one upstream and previously described and the other downstream and apparently novel. The haplotype comprising these SNPs was strongly associated with AGA (P = 3.75 × 10(-5)) in 1195 men. We also replicated association with a recently reported non-coding region on chromosome 20 and found that its association with AGA was less strong and independent of that of AR. Our results will help focus future efforts to further define AGA genetic risk.


Asunto(s)
Alopecia/genética , Polimorfismo de Nucleótido Simple/genética , Receptores Androgénicos/genética , Adolescente , Adulto , Anciano , Alopecia/diagnóstico , Cromosomas Humanos Par 20/genética , Epistasis Genética/genética , Frecuencia de los Genes/genética , Genes Ligados a X/genética , Genotipo , Humanos , Desequilibrio de Ligamiento/genética , Masculino , Persona de Mediana Edad , Receptor Xedar/genética , Adulto Joven
20.
Nature ; 424(6950): 793-6, 2003 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-12917689

RESUMEN

Familial cylindromatosis is an autosomal dominant predisposition to tumours of skin appendages called cylindromas. Familial cylindromatosis is caused by mutations in a gene encoding the CYLD protein of previously unknown function. Here we show that CYLD is a deubiquitinating enzyme that negatively regulates activation of the transcription factor NF-kappaB by specific tumour-necrosis factor receptors (TNFRs). Loss of the deubiquitinating activity of CYLD correlates with tumorigenesis. CYLD inhibits activation of NF-kappaB by the TNFR family members CD40, XEDAR and EDAR in a manner that depends on the deubiquitinating activity of CYLD. Downregulation of CYLD by RNA-mediated interference augments both basal and CD40-mediated activation of NF-kappaB. The inhibition of NF-kappaB activation by CYLD is mediated, at least in part, by the deubiquitination and inactivation of TNFR-associated factor 2 (TRAF2) and, to a lesser extent, TRAF6. These results indicate that CYLD is a negative regulator of the cytokine-mediated activation of NF-kappaB that is required for appropriate cellular homeostasis of skin appendages.


Asunto(s)
FN-kappa B/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina/metabolismo , Antígenos CD40/metabolismo , Línea Celular , Enzima Desubiquitinante CYLD , Receptor Edar , Humanos , Quinasa I-kappa B , Proteínas de la Membrana/metabolismo , Mutación , FN-kappa B/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Interferencia de ARN , Receptores de la Ectodisplasina , Factor 2 Asociado a Receptor de TNF , Factor 6 Asociado a Receptor de TNF , Proteínas Supresoras de Tumor/genética , Receptor Xedar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA