Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 355
Filtrar
1.
J Am Heart Assoc ; 12(8): e028625, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37042257

RESUMEN

Background Differences in the clinical course of heritable thoracic aortic disease based on the disease-causing gene have not been fully evaluated. To clarify the clinical relevance of causative genes in heritable thoracic aortic disease, we assessed the clinical course of patients categorized based on genetic diagnosis. Methods and Results We investigated cardiovascular events and mortality in 518 genetically diagnosed patients in 4 groups: Group 1, FBN1 (n=344); Group 2, TGFBR1, TGFBR2, SMAD3, or TGFB2 (n=74); Group 3, COL3A1 (n=60); and Group 4, ACTA2 or MYH11 (n=40). The median age at the first cardiovascular event ranged from 30.0 to 35.5 years (P=0.36). Patients with gene variants related to transforming growth factor-ß signaling had a significantly higher rate of subsequent events than those with FBN1 variants (adjusted hazard ratio, 2.33 [95% CI, 1.60-3.38]; P<0.001). Regarding the incidence of aortic dissection, there were no significant differences among the 4 groups in male patients (36.3%, 34.3%, 21.4%, and 54.2%, respectively; P=0.06). Female patients with COL3A1 variants had a significantly lower incidence than female patients in the other 3 groups (34.2%, 59.0%, 3.1%, and 43.8%, respectively; P<0.001). Conclusions Gene variants related to transforming growth factor-ß signaling are associated with a higher incidence of subsequent cardiovascular events than FBN1 variants. COL3A1 variants might be related to a lower incidence of aortic dissection than other gene variants in women only. Identifying the genetic background of patients with heritable thoracic aortic disease is important for determining appropriate treatment.


Asunto(s)
Aneurisma de la Aorta Torácica , Disección Aórtica , Humanos , Masculino , Femenino , Adulto , Receptores de Factores de Crecimiento Transformadores beta/genética , Disección Aórtica/diagnóstico , Disección Aórtica/genética , Transducción de Señal/genética , Factores de Crecimiento Transformadores/genética , Progresión de la Enfermedad , Aneurisma de la Aorta Torácica/diagnóstico , Aneurisma de la Aorta Torácica/genética , Mutación
2.
J Obstet Gynaecol Res ; 49(1): 232-242, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36396030

RESUMEN

BACKGROUND: Endometrial dysfunction is closely correlated with the development of multiple severe gynecological disorders including intrauterine adhesion. Accumulating evidence supports that some long non-coding RNAs (lncRNAs) have peptide-coding potential. In this text, the peptide-coding ability of lncRNA SNHG6 was examined. Also, the effects of an SNHG6-encoded peptide on the viability and migration of human endometrial stromal cells (hESCs) and human endometrial epithelial cells (hEECs) and related molecular mechanisms were explored. METHODS: The peptide-encoding potential of SNHG6 was predicted by FuncPEP and getorf databases and validated by western blot assay. Cell viability was tested by cell counting kit-8 assay. Cell migratory ability was examined by wound healing and transwell migration assays. Protein levels of genes were measured by western blot assay. RESULTS: Prediction analysis suggested that SNHG6 had the potential peptide-coding ability and multiple open-reading frames (ORFs). Western blot validated that SNHG6 ORF#1 and ORF#2 could translate into short peptides. SNHG6 ORF#2 overexpression facilitated cell migration and epithelial-mesenchymal transition (EMT) in hESCs and hEECs, while these effects were abrogated by transforming growth factor-beta (TGF-ß)/SMAD signaling inhibitor GW788388. Moreover, GW788388 inhibited the increase of p-SMAD2 and p-SMAD3 levels induced by SNHG6 ORF#2 in hESCs. SNHG6 ORF#2-encoded peptide did not influence endometrial stromal and epithelial cell viability. CONCLUSIONS: LncRNA SNHG6 ORF#1 and ORF#2 could translate into small peptides and SNHG6 ORF#2 overexpression promoted cell migration and EMT by activating the TGF-ß/SMAD pathway in hESCs and hEECs, suggesting the potential roles of SNHG6-encoded peptides in the development of endometrial stromal and epithelial cells and related gynecological diseases.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transición Epitelial-Mesenquimal/genética , ARN Nucleolar Pequeño/farmacología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento Transformador beta/farmacología , Transducción de Señal , Movimiento Celular/genética , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/farmacología
3.
J Thorac Oncol ; 18(4): 499-515, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36535627

RESUMEN

INTRODUCTION: Targeted therapies require life-long treatment, as drug discontinuation invariably leads to tumor recurrence. Recurrence is mainly driven by minor subpopulations of drug-tolerant persister (DTP) cells that survive the cytotoxic drug effect. In lung cancer, DTP studies have mainly been conducted with cell line models. METHODS: We conducted an in vivo DTP study using a lung adenocarcinoma patient-derived xenograft tumor driven by an EGFR mutation. Daily treatment of tumor-bearing mice for 5 to 6 weeks with the EGFR inhibitor erlotinib markedly shrunk tumors and generated DTPs, which were analyzed by whole exome, bulk population transcriptome, and single-cell RNA sequencing. RESULTS: The DTP tumors maintained the genomic clonal architecture of untreated baseline (BL) tumors but had reduced proliferation. Single-cell RNA sequencing identified a rare (approximately 4%) subpopulation of BL cells (DTP-like) with transcriptomic similarity to DTP cells and intermediate activity of pathways that are up-regulated in DTPs. Furthermore, the predominant transforming growth factor-ß activated cancer-associated fibroblast (CAF) population in BL tumors was replaced by a CAF population enriched for IL6 production. In vitro experiments indicate that these populations interconvert depending on the levels of transforming growth factor-ß versus NF-κB signaling, which is modulated by tyrosine kinase inhibitor presence. The DTPs had signs of increased NF-κB and STAT3 signaling, which may promote their survival. CONCLUSIONS: The DTPs may arise from a specific preexisting subpopulation of cancer cells with partial activation of specific drug resistance pathways. Tyrosine kinase inhibitor treatment induces DTPs revealing greater activation of these pathways while converting the major preexisting CAF population into a new state that may further promote DTP survival.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Animales , Ratones , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Transcriptoma , FN-kappa B/genética , FN-kappa B/metabolismo , FN-kappa B/farmacología , Xenoinjertos , Receptores ErbB/metabolismo , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de la Célula Individual , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/farmacología , Factores de Crecimiento Transformadores/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Mutación
4.
Ren Fail ; 44(1): 1819-1832, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36299239

RESUMEN

BACKGROUND/AIM: Uremic cardiomyopathy (UCM) is a characteristic cardiac pathology that is commonly found in patients with chronic kidney disease. This study dissected the mechanism of SPI1 in myocardial fibrosis and inflammation induced by UCM through S100A8/A9. METHODS: An UCM rat model was established, followed by qRT-PCR and western blot analyses of SPI1 and S100A8/A9 expression in myocardial tissues. After alterations of SPI1 and S100A8/A9 expression in UCM rats, the blood specimens were harvested from the cardiac apex of rats. The levels of creatine phosphokinase-MB (CK-MB), blood creatinine, blood urea nitrogen (BUN), and inflammatory cytokines (interleukin [IL]-6, IL-1ß, and tumor necrosis factor-α [TNF-α]) were examined in the collected blood. Collagen fibrosis was assessed by Masson staining. The expression of fibrosis markers [transforming growth factor (TGF)-ß1, α-smooth muscle actin (SMA), Collagen 4a1, and Fibronectin], IL-6, IL-1ß, and TNF-α was measured in myocardial tissues. Chromatin immunoprecipitation and dual-luciferase reporter gene assays were conducted to test the binding relationship between SPI1 and S100A8/A9. RESULTS: S100A8/A9 and SPI1 were highly expressed in the myocardial tissues of UCM rats. Mechanistically, SPI1 bound to the promoter of S100A8/A9 to facilitate S100A8/A9 transcription. S100A8/A9 or SPI1 knockdown reduced myocardial fibrosis and inflammation and the levels of CK-MB, blood creatinine, and BUN, as well as the expression of TGF-ß1, α-SMA, Collagen 4a1, Fibronectin, IL-6, TNF-α, and IL-1ß in UCM rats. CONCLUSION: SPI1 knockdown diminished S100A8/A9 transcription, thus suppressing myocardial fibrosis and inflammation caused by UCM.


Asunto(s)
Calgranulina A , Calgranulina B , Cardiomiopatías , Animales , Ratas , Actinas/metabolismo , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/prevención & control , Creatina Quinasa , Creatinina , Citocinas/metabolismo , Regulación hacia Abajo , Fibronectinas/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Islas Genómicas , Inflamación/genética , Inflamación/metabolismo , Interleucina-6/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Uremia/complicaciones , Uremia/genética , Uremia/metabolismo
5.
Molecules ; 27(18)2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36144752

RESUMEN

The transforming growth factor-ß (TGF-ß) superfamily encodes a large group of proteins, including TGF-ß isoforms, bone morphogenetic proteins and activins that act through conserved cell-surface receptors and signaling co-receptors. TGF-ß signaling in insects controls physiological events, including growth, development, diapause, caste determination and metamorphosis. In this study, we used the red flour beetle, Tribolium castaneum, as a model species to investigate the role of the type I TGF-ß receptor, saxophone (Sax), in mediating development. Developmental and tissue-specific expression profiles indicated Sax is constitutively expressed during development with lower expression in 19- and 20-day (6th instar) larvae. RNAi knockdown of Sax in 19-day larvae prolonged developmental duration from larvae to pupae and significantly decreased pupation and adult eclosion in a dose-dependent manner. At 50 ng dsSax/larva, Sax knockdown led to an 84.4% pupation rate and 46.3% adult emergence rate. At 100 ng and 200 ng dsSax/larva, pupation was down to 75.6% and 50%, respectively, with 0% adult emergence following treatments with both doses. These phenotypes were similar to those following knockdowns of 20-hydroxyecdysone (20E) receptor genes, ecdysone receptor (EcR) or ultraspiracle protein (USP). Expression of 20E biosynthesis genes disembodied and spookier, 20E receptor genes EcR and USP, and 20E downstream genes BrC and E75, were suppressed after the Sax knockdown. Topical application of 20E on larvae treated with dsSax partially rescued the dsSax-driven defects. We can infer that the TGF-ß receptor gene Sax influences larval-pupal-adult development via 20E signaling in T. castaneum.


Asunto(s)
Tribolium , Activinas/genética , Activinas/metabolismo , Animales , Ecdisterona , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Larva , Isoformas de Proteínas/metabolismo , Pupa/genética , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
6.
Cell Tissue Res ; 390(3): 465-489, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36098854

RESUMEN

Overexposure to transforming growth factor b1 (TGF-ß1) induces myofibroblastic differentiation of mesenchymal stem cells (MSCs), which could be attenuated by myeloid-derived suppressor cell (MDSC) supernatant. However, the promyofibroblastic effects of TGF-ß1 and the antimyofibroblastic effects of MDSC supernatant in MSCs have not been fully elucidated. To further clarify the latent mechanism and identify underlying therapeutic targets, we used an integrative strategy combining transcriptomics and metabolomics. Bone marrow MSCs were collected 24 h following TGF-ß1 and MDSC supernatant treatment for RNA sequencing and untargeted metabolomic analysis. The integrated data were then analyzed to identify significant gene-metabolite correlations. Differentially expressed genes (DEGs) and differentially expressed metabolites (DEMs) were assessed by Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses for exploring the mechanisms of myofibroblastic differentiation of MSCs. The integration of transcriptomic and metabolomic data highlighted significantly coordinated changes in glycolysis/gluconeogenesis and purine metabolism following TGF-ß1 and MDSC supernatant treatment. By combining transcriptomic and metabolomic analyses, this study showed that glycolysis/gluconeogenesis and purine metabolism were essential for the myofibroblastic differentiation of MSCs and may serve as promising targets for mechanistic research and clinical practice in the treatment of fibrosis by MDSC supernatant.


Asunto(s)
Células Madre Mesenquimatosas , Células Supresoras de Origen Mieloide , Miofibroblastos , Diferenciación Celular , Células Supresoras de Origen Mieloide/metabolismo , Purinas/metabolismo , Purinas/farmacología , Transcriptoma/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/farmacología , Miofibroblastos/citología
7.
Cell Mol Biol Lett ; 27(1): 79, 2022 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-36138344

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) play crucial roles in the development of hepatocellular carcinoma (HCC). Hsa-microRNA-27b-3p (hsa-miR-27b) is involved in the formation and progression of various cancers, but its role and clinical value in HCC remain unclear. METHODS: The expression of hsa-miR-27b in HCC was examined by quantitative real-time PCR (qRT-PCR) and in situ hybridization (ISH) assays of clinical samples. Cell Counting Kit-8 assays (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays, Transwell assays, filamentous actin (F-actin) staining and western blot analyses were used to determine the effects of hsa-miR-27b on HCC cells in vitro. Subcutaneous xenograft and lung metastatic animal experiments were conducted to verify the role of hsa-miR-27b in HCC in vivo. In silico prediction, qRT-PCR, western blot, anti-Argonaute 2 (AGO2) RNA immunoprecipitation (RIP) and dual luciferase reporter assays were applied to identify the target genes of hsa-miR-27b. To detect the impacts of hsa-miR-27b on nuclear factor kappa B (NF-кB) signalling cascades mediated by transforming growth factor-activated kinase-binding protein 3 (TAB3), we performed qRT-PCR, western blot assays, immunofluorescence staining, immunohistochemistry (IHC) and dual-luciferase reporter assays. Recombinant oncolytic adenovirus (OncoAd) overexpressing hsa-miR-27b was constructed to detect their therapeutic value in HCC. RESULTS: The expression of hsa-miR-27b was lower in HCC than in adjacent non-tumourous tissues (ANTs), and the reduced expression of hsa-miR-27b was associated with worse outcomes in patients with HCC. Hsa-miR-27b significantly inhibited the proliferation, migration, invasion, subcutaneous tumour growth and lung metastasis of HCC cells. The suppression of hsa-miR-27b promoted the nuclear translocation of NF-κB by upregulating TAB3 expression. TAB3 was highly expressed in HCC compared with ANTs and was negatively correlated with the expression of hsa-miR-27b. The impaired cell proliferation, migration and invasion by hsa-miR-27b overexpression were recovered by ectopic expression of TAB3. Recombinant OncoAd with overexpression of hsa-miR-27b induced anti-tumour activity compared with that induced by negative control (NC) OncoAd in vivo and in vitro. CONCLUSIONS: By targeting TAB3, hsa-miR-27b acted as a tumour suppressor by inactivating the NF-кB pathway in HCC in vitro and in vivo, indicating its therapeutic value against HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , Actinas/genética , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/patología , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
8.
Mol Ther ; 30(10): 3257-3269, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-35780298

RESUMEN

Previously we found that inhibitor of differentiation 3 (Id3) gene, a transcriptional repressor, efficiently inhibits corneal keratocyte differentiation to myofibroblasts in vitro. This study evaluated the potential of adeno-associated virus 5 (AAV5)-mediated Id3 gene therapy to treat corneal scarring using an established rabbit in vivo disease model. Corneal scarring/fibrosis in rabbit eyes was induced by alkali trauma, and 24 h thereafter corneas were administered with either balanced salt solution AAV5-naked vector, or AAV5-Id3 vector (n = 6/group) via an optimized reported method. Therapeutic effects of AAV5-Id3 gene therapy on corneal pathology and ocular health were evaluated with clinical, histological, and molecular techniques. Localized AAV5-Id3 gene therapy significantly inhibited corneal fibrosis/haze clinically from 2.7 to 0.7 on the Fantes scale in live animals (AAV5-naked versus AAV5-Id3; p < 0.001). Furthermore, AAV5-Id3 treatment significantly reduced profibrotic gene mRNA levels: α-smooth muscle actin (α-SMA) (2.8-fold; p < 0.001), fibronectin (3.2-fold; p < 0.001), collagen I (0.8-fold; p < 0.001), and collagen III (1.4-fold; p < 0.001), as well as protein levels of α-SMA (23.8%; p < 0.001) and collagens (1.8-fold; p < 0.001). The anti-fibrotic activity of AAV5-Id3 is attributed to reduced myofibroblast formation by disrupting the binding of E-box proteins to the promoter of α-SMA, a transforming growth factor-ß signaling downstream target gene. In conclusion, these results indicate that localized AAV5-Id3 delivery in stroma caused no clinically relevant ocular symptoms or corneal cellular toxicity in the rabbit eyes.


Asunto(s)
Enfermedades de la Córnea , Lesiones de la Cornea , Opacidad de la Córnea , Actinas/genética , Álcalis , Animales , Cicatriz/patología , Cicatriz/terapia , Córnea , Enfermedades de la Córnea/genética , Enfermedades de la Córnea/terapia , Lesiones de la Cornea/patología , Lesiones de la Cornea/terapia , Opacidad de la Córnea/patología , Opacidad de la Córnea/terapia , Dependovirus , Fibronectinas/genética , Fibrosis , Terapia Genética/métodos , ARN Mensajero , Conejos , Factores de Crecimiento Transformadores/genética
9.
Cell Cycle ; 21(21): 2298-2308, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35856444

RESUMEN

Gastric cancer is among the most common gastrointestinal malignancies. Recent studies have suggested that bone morphogenetic protein-2 (BMP2) is related to the development and progression of various cancers. Meanwhile, evidence suggests that BMP2 might lead to epigenetic changes in gastric cancer. Thus, we investigated whether BMP2 plays a role in the development of gastric cancer via epigenetic regulation. Cell viability, colony formation, and cell cycle assays were performed to assess the effect of recombinant human BMP2 (rhBMP2) in gastric cancer cells. LDN-193189 and Noggins were used as antagonists of the canonical BMP-SMAD signaling pathway. The protein levels were determined using a western blot analysis. Lentiviral vectors with EZH2 shRNA or EZH2 overexpression were used to mediate the role of EZH2 and the relationship between BMP2 and EZH2 in gastric cancer. We found that rhBMP2 inhibits cell proliferation by arresting the cell cycle in HGC-27 and SNU-216 gastric cancer cells. Neither LDN-193189 nor Noggins, antagonists of the canonical BMP-SMAD signaling pathway, can reverse the effect of rhBMP2 on gastric cancer. Molecularly, rhBMP2 downregulates the expression of EZH2 and H3K27me3, leading to increases in P16 and P21 and decreases in CDK2, CDK4, and CDK6. Altogether, in this study, we demonstrate that BMP2 serves as a tumor suppressor in gastric cancer cells by downregulating EZH2 and H3K27me3 through the non-SMAD BMP pathway, suggesting that BMP2 might be a new therapeutic target for gastric cancer treatment. Abbreviations: BMP: bone morphogenetic protein; TGF-ß: transforming growth factor-beta; EZH2: enhancer of zeste homolog 2; H3K27me3: trimethylation histone H3 lysine 27; HRECs: human retinal endothelial cells; PcG: polycomb group; PRC: polycomb repressive complexes.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Neoplasias Gástricas , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , ARN Interferente Pequeño , Neoplasias Gástricas/genética , Factor de Crecimiento Transformador beta/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
10.
Gene ; 841: 146763, 2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-35907564

RESUMEN

Recently, He et al. performed a meta-analysis to interpret the association between transforming growth factor-ß (TGF-ß) gene polymorphisms and chronic obstructive pulmonary disease (COPD) risk. However, we would like to comment on some debatable points shown in this meta-analysis.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica , Factor de Crecimiento Transformador beta , Predisposición Genética a la Enfermedad , Humanos , Polimorfismo Genético , Enfermedad Pulmonar Obstructiva Crónica/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/genética , Factores de Crecimiento Transformadores/genética
11.
J Biochem Mol Toxicol ; 36(9): e23144, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35730126

RESUMEN

Posterior capsular opacification (PCO) is the major complication after cataract surgery and can result in secondary vision loss. Circular RNAs (circRNAs) are reported to play critical regulatory roles in multiple cell biological processes. The most common working mechanism of circRNAs is by acting as microRNA sponges. Here, we analyzed the role and mechanism of circRNA RNA polymerase III subunit A (POLR3A) in PCO. Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Cell motility was assessed by transwell and wound healing assays. Dual-luciferase reporter and RNA-pull-down assays were performed to verify the interaction between microRNA-31 (miR-31) and circ-POLR3A or thioredoxin interacting protein (TXNIP). PCO cell model was established by treating SRA01/04 cells with transforming growth factor-ß2 (TGF-ß2). We found that TGF-ß2 enhanced SRA01/04 cell viability, migration, and invasion abilities. Circ-POLR3A expression was upregulated in PCO tissues and TGF-ß2-induced SRA01/04 cells. TGF-ß2 promoted the viability and motility of SRA01/04 cells largely by upregulating circ-POLR3A. Circ-POLR3A negatively regulated the miR-31 level by directly interacting with it. Circ-POLR3A absence-induced influences in TGF-ß2-induced SRA01/04 cells were partly reversed by silencing miR-31. miR-31 is directly bound to the 3'-untranslated region of TXNIP. TXNIP overexpression largely attenuated miR-31 overexpression-mediated effects in TGF-ß2-induced SRA01/04 cells. Circ-POLR3A could elevate the protein expression of TXNIP by sponging miR-31. Exosomes were involved in mediating the delivery of circ-POLR3A in SRA01/04 cells. In conclusion, circ-POLR3A contributed to TGF-ß2-induced promotion of cell viability, migration, and invasion of SRA01/04 cells by targeting miR-31/TXNIP axis.


Asunto(s)
Opacificación Capsular , MicroARNs , Regiones no Traducidas 3' , Opacificación Capsular/genética , Opacificación Capsular/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Humanos , MicroARNs/metabolismo , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , ARN Polimerasa III/farmacología , ARN Circular/genética , Tiorredoxinas , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta2/farmacología , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/farmacología
12.
Cells ; 11(7)2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35406794

RESUMEN

During osteoarthritis (OA), hypertrophy-like chondrocytes contribute to the disease process. TGF-ß's signaling pathways can contribute to a hypertrophy(-like) phenotype in chondrocytes, especially at high doses of TGF-ß. In this study, we examine which transcription factors (TFs) are activated and involved in TGF-ß-dependent induction of a hypertrophy-like phenotype in human OA chondrocytes. We found that TGF-ß, at levels found in synovial fluid in OA patients, induces hypertrophic differentiation, as characterized by increased expression of RUNX2, COL10A1, COL1A1, VEGFA and IHH. Using luciferase-based TF activity assays, we observed that the expression of these hypertrophy genes positively correlated to SMAD3:4, STAT3 and AP1 activity. Blocking these TFs using specific inhibitors for ALK-5-induced SMAD signaling (5 µM SB-505124), JAK-STAT signaling (1 µM Tofacitinib) and JNK signaling (10 µM SP-600125) led to the striking observation that only SB-505124 repressed the expression of hypertrophy factors in TGF-ß-stimulated chondrocytes. Therefore, we conclude that ALK5 kinase activity is essential for TGF-ß-induced expression of crucial hypertrophy factors in chondrocytes.


Asunto(s)
Condrocitos , Osteoartritis , Condrocitos/metabolismo , Humanos , Hipertrofia/metabolismo , Osteoartritis/genética , Osteoartritis/metabolismo , Fenotipo , Factor de Crecimiento Transformador beta/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
13.
Front Immunol ; 13: 836226, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309318

RESUMEN

The transforming growth factor-ßs (TGF-ßs) are multifunctional cytokines capable of regulating a wide range of cellular behaviors and play a key role in maintaining the homeostasis of the immune system. The TGF-ß subfamily, which is only present in deuterostomes, expands from a single gene in invertebrates to multiple members in jawed vertebrates. However, the evolutionary processes of the TGF-ß subfamily in vertebrates still lack sufficient elucidation. In this study, the TGF-ß homologs are identified at the genome-wide level in the reissner lamprey (Lethenteron reissneri), the sea lamprey (Petromyzon marinus), and the Japanese lamprey (Lampetra japonica), which are the extant representatives of jawless vertebrates with a history of more than 350 million years. The molecular evolutionary analyses reveal that the lamprey TGF-ß subfamily contains two members representing ancestors of TGF-ß2 and 3 in vertebrates, respectively, but TGF-ß1 is absent. The transcriptional expression patterns show that the lamprey TGF-ß2 may play a central regulatory role in the innate immune response of the lamprey since it exhibits a more rapid and significant upregulation of expression than TGF-ß3 during lipopolysaccharide stimuli. The incorporation of BrdU assay reveals that the lamprey TGF-ß2 recombinant protein exerts the bipolar regulation on the proliferation of the supraneural myeloid body cells (SMB cells) in the quiescent and LPS-activated state, while plays an inhibitory role in the proliferation of quiescent and activated leukocytes in lampreys. Furthermore, caspase-3/7 activity analysis indicates that the lamprey TGF-ß2 protects SMB cells from apoptosis after serum deprivation, in contrast to promoting apoptosis of leukocytes. Our composite results offer valuable clues to the origin and evolution of the TGF-ß subfamily and imply that TGF-ßs are among the most ancestral immune regulators in vertebrates.


Asunto(s)
Petromyzon , Factor de Crecimiento Transformador beta2 , Animales , Evolución Molecular , Lipopolisacáridos/farmacología , Filogenia , Factor de Crecimiento Transformador beta2/genética , Factores de Crecimiento Transformadores/genética , Vertebrados
14.
Ophthalmic Genet ; 43(4): 530-533, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35315300

RESUMEN

PURPOSE: To provide the initial confirmation of the c.1772C>T (p.Ser591Phe) mutation in the transforming growth factor-ß-induced (TGFBI) gene as being associated with variant lattice corneal dystrophy (LCD). METHODS: Ophthalmologic examination of the proband was performed with slit lamp biomicroscopy. Saliva was collected as a source of DNA for screening all 17 exons of TGFBI, after which three family members were selectively screened for variants in exon 13. Rosetta-based structure prediction was used to calculate changes in TGFBI protein (TGFBIp) stability secondary to the c.1772C>T (p.Ser591Phe) missense mutation. RESULTS: Slit lamp examination of the 38-year-old proband revealed a clear cornea right eye and unilateral, discrete, and branching lattice lines in the anterior and mid-stroma of the central cornea left eye. Screening of TGFBI in the proband revealed a heterozygous missense mutation in exon 13 (c.1772C>T (p.Ser591Phe)) that was also identified in her affected mother but not in her brother or maternal grandmother. Calculated energy change in Rosetta (ΔΔG) for the TGFBIp variant p.Ser591Phe was 23.5, indicating a thermodynamic destabilization resulting from energetic frustration. CONCLUSIONS: The p.Ser591Phe mutation in TGFBI is associated with an unilateral variant of LCD. Rosetta-predicted stability changes indicate that the p.Ser591Phe variant is destabilizing, which is consistent with other observations for LCD-causing mutations.


Asunto(s)
Neuropatías Amiloides Familiares , Distrofias Hereditarias de la Córnea , Proteínas de la Matriz Extracelular , Factores de Crecimiento Transformadores , Adulto , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/genética , Análisis Mutacional de ADN , Proteínas de la Matriz Extracelular/genética , Femenino , Humanos , Masculino , Mutación , Mutación Missense , Linaje , Factor de Crecimiento Transformador beta , Factores de Crecimiento Transformadores/genética
15.
Int J Biol Macromol ; 206: 435-452, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35202639

RESUMEN

Transforming growth factor-beta (TGF-ß) is a member of a family of secreted cytokines with vital biological functions in cells. The abnormal expression of TGF-ß signaling is a common finding in pathological conditions, particularly cancer. Prostate cancer (PCa) is one of the leading causes of death among men. Several genetic and epigenetic alterations can result in PCa development, and govern its progression. The present review attempts to shed some light on the role of TGF-ß signaling in PCa. TGF-ß signaling can either stimulate or inhibit proliferation and viability of PCa cells, depending on the context. The metastasis of PCa cells is increased by TGF-ß signaling via induction of EMT and MMPs. Furthermore, TGF-ß signaling can induce drug resistance of PCa cells, and can lead to immune evasion via reducing the anti-tumor activity of cytotoxic T cells and stimulating regulatory T cells. Upstream mediators such as microRNAs and lncRNAs, can regulate TGF-ß signaling in PCa. Furthermore, some pharmacological compounds such as thymoquinone and valproic acid can suppress TGF-ß signaling for PCa therapy. TGF-ß over-expression is associated with poor prognosis in PCa patients. Furthermore, TGF-ß up-regulation before prostatectomy is associated with recurrence of PCa. Overall, current review discusses role of TGF-ß signaling in proliferation, metastasis and therapy response of PCa cells and in order to improve knowledge towards its regulation, upstream mediators of TGF-ß such as non-coding RNAs are described. Finally, TGF-ß regulation and its clinical application are discussed.


Asunto(s)
Neoplasias de la Próstata , Factor de Crecimiento Transformador beta , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
16.
Ir J Med Sci ; 191(5): 2029-2033, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34792732

RESUMEN

BACKGROUND: Gestational diabetes mellitus (GDM) reflects a deficiency in the relative need for insulin during pregnancy, as well as temporary metabolic stress in the placenta and fetus. Our study aimed to research the potential diagnostic value of transforming growth factor-beta-induced protein ig-h3 (TGFBI) and transforming growth factor beta-2 proprotein (TGFB2) for GDM patients. METHODS: Online database Gene Expression Omnibus (GEO) was used to screen for different expressed genes (DEGs) associated with GDM. Meanwhile, KEGG and GO were used to analyze the molecular functions as well as pathways of enriched DEGs. One hundred ten pregnant women diagnosed with GDM and 110 healthy controls were enrolled, of whose placenta and fasting venous blood samples were collected. mRNA expression levels were determined by real-time quantitative polymerase chain reaction (RT-qPCR), and fasting blood glucose (FBG) was measured by the clinical lab of hospital. Furthermore, receiver operating characteristics curve (ROC) analysis was performed to evaluate the sensitivity and specificity of detection indexed in the placenta and plasma of GDM patients. Finally, Pearson and Spearman analysis was used for the correlation analysis. RESULTS: After GEO data analysis, TGFBI and TGFB2 were identified as the most significantly up-regulated genes of GDM. TGFBI and TGFB2 expressions in placenta and plasma samples of GDM patients were in line with bioinformatic analysis. Meanwhile, the area under the curve (AUC) of TGFBI in the placenta and plasma for the diagnosis of GDM were 0.8783 (95% CI, 0.8281 to 0.9284) and 0.7832 (95% CI, 0.7215 to 0.8449) while for TGFB2 were 0.9225 (95% CI, 0.8829 to 0.9621) and 0.8961 (95% CI, 0.8526 to 0.9396). Besides, levels of TGFBI along with TGFB2 in the placenta were positively correlated with that in the plasma of GDM patients. Furthermore, both TGFBI and TGFB2 expressions in the plasma were positively correlated with FBG levels of the GDM patients. CONCLUSIONS: TGFBI and TGFB2 were up-regulated in the placenta and plasma of GDM patients, and TGFBI and TGFB2 in the plasma are potent to be diagnostic markers for the GDM.


Asunto(s)
Diabetes Gestacional , Glucemia/metabolismo , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/genética , Femenino , Humanos , Insulina , Embarazo , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Regulación hacia Arriba
17.
Ann Surg Oncol ; 29(5): 2944-2956, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34855064

RESUMEN

BACKGROUND: Transient receptor potential vanilloid 2 (TRPV2) is a highly Ca2+-permeable ion channel that is involved in a number of cellular processes. It is expressed in various human cancers; however, the role of TRPV2 in gastric cancer (GC) remains poorly understood. METHODS: TRPV2 gene expression was knocked down in GC cell lines by small-interfering RNA (siRNA), and the biological roles of TRPV2 in the proliferation, migration, and invasion of GC cells were then investigated. The gene expression profile of GC was elucidated using a microarray analysis. TRPV2 expression in tumor tissue sections was analyzed by immunohistochemistry. RESULTS: The migration and invasion abilities of GC cells were inhibited by the knockdown of TRPV2. Moreover, the microarray assay revealed that TRPV2 was associated with the transforming growth factor (TGF)-ß signaling pathway. Immunohistochemical staining showed that the strong expression of TRPV2 correlated with lymphatic invasion, venous invasion, pathological T (pT), pathological N (pN), and a poor prognosis in GC patients. CONCLUSIONS: TRPV2 appeared to promote tumor migration and invasion via the TGF-ß signaling pathway, and the strong expression of TRPV2 was associated with a worse prognosis in GC patients.


Asunto(s)
Neoplasias Gástricas , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica/genética , ARN Interferente Pequeño , Transducción de Señal , Neoplasias Gástricas/patología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
18.
DNA Cell Biol ; 40(7): 988-997, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34061659

RESUMEN

Primary open-angle glaucoma (POAG) is tightly related with extracellular matrix (ECM) remodeling of human trabecular meshwork cells (HTMCs). Transforming growth factor-ß2 (TGF-ß2) can induce ECM remodeling. The aim of the study was to investigate the microRNAs (miRNAs) expression changes of extracellular vesicles (EVs) derived from HTMCs treated with TGF-ß2. EVs were isolated from HTMCs supernatant cultured for 24 h with TGF-ß2. The morphology of EVs pellets was examined by transmission electron microscopy. Nanoparticle tracking analysis used to demonstrate the particle size distribution. Total EVs RNAs were extracted for subsequent miRNA gene chip analysis to investigate differentially expressed miRNAs between the controls and treatment cells. Gene Ontology (GO) annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to predict potential target and validate possible functions of the miRNAs. There were 23 miRNAs upregulated and 3 miRNAs downregulated and 469,102, and 94 GO terms involved in biological processes, cellular components, and molecular function for the possible functions of the 26 miRNAs. These findings indicate that TGF-ß2 may alter EVs miRNAs expression to participate in the pathogenesis of POAG. They may provide significant information for potential biomarkers for POAG diagnosis and treatment.


Asunto(s)
Vesículas Extracelulares/genética , Malla Trabecular/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Células Cultivadas , China , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/metabolismo , Humanos , MicroARNs/genética , Cultivo Primario de Células , Malla Trabecular/fisiología , Transcriptoma/genética , Factor de Crecimiento Transformador beta2/genética , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
19.
Chin Med J (Engl) ; 132(16): 1942-1950, 2019 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-31365430

RESUMEN

BACKGROUND: Henoch-Schonlein purpura nephritis (HSPN) is a very common secondary kidney disease of childhood. Its pathogenesis and the treatment mechanism of glucocorticoid have not been fully elucidated. The aim of this study was to determine the relationship between p300 and the pathogenesis, glucocorticoid therapy in mice with HSPN, respectively. METHODS: Forty-eight C57BL/6N male mice, weighing 18 to 20 g, were selected (3-4 weeks old, n = 8 per group). The mice in the normal control group (Group I) were given normal solvent and the HSPN model group (Group II) were given sensitizing drugs. The mice in Group III were injected intraperitoneally with dexamethasone after being given sensitizing drugs. Meanwhile, mice in Groups IV, V and VI with conditional knockout of p300 were also given normal solvent, sensitizing drugs and dexamethasone.The levels of serum IgA, creatinine, and circulating immune complex (CIC) concentrations, 24 h urinary protein and urinary erythrocyte in C57 wild mice, and p300 conditional knockout mice in each group were measured. The expression of p300 in renal tissues and the expression of glucocorticoid receptor (GR) α and ß, transforming growth factor (TGF)-ß1, and activator protein (AP)-1 after dexamethasone treatment were determined by real-time polymerase chain reaction and Western blotting. RESULTS: Compared with the normal solvent control group (Group I), the expression of p300 mRNA in the model group (Group II) was significantly up-regulated. Western blotting further confirmed the result. Urinary erythrocyte count, 24 h urinary protein quantification, serum IgA, CIC, and renal pathologic score in Group V were distinctly decreased compared with non-knockout mice in Group II (9.7 ±â€Š3.8 per high-power field [/HP] vs. 18.7 ±â€Š6.2/HP, t = 1.828, P = 0.043; 0.18 ±â€Š0.06 g/24 h vs. 0.36 ±â€Š0.08 g/24 h, t = 1.837, P = 0.042; 18.78 ±â€Š0.85 mg/mL vs. 38.46 ±â€Š0.46 mg/mL, t = 1.925, P = 0.038; 0.80 ±â€Š0.27 µg/mL vs. 1.64 ±â€Š0.47 µg/mL, t = 1.892, P = 0.041; 7.0 ±â€Š0.5 vs. 18.0 ±â€Š0.5, t = 1.908, P = 0.039). Compared with non-knockout mice (Group III), the level of urinary erythrocyte count and serum IgA in knockout mice (Group VI) increased significantly after treatment with dexamethasone (3.7 ±â€Š0.6/HP vs. 9.2 ±â€Š3.5/HP, t = 2.186, P = 0.024; 12.38 ±â€Š0.26 mg/mL vs. 27.85 ±â€Š0.65 mg/mL, t = 1.852, P = 0.041). The expression level of GRα was considerably increased in the knockout group after dexamethasone treatment compared with non-knockout mice in mRNA and protein level (t = 2.085, P = 0.026; t = 1.928, P = 0.035), but there was no statistically significant difference in the expression level of GRß between condition knockout and non-knockout mice (t = 0.059, P = 0.087; t = 0.038, P = 1.12). Furthermore, the expression levels of glucocorticoid resistance genes (AP-1 and TGF-ß1) were notably increased after p300 knockout compared with non-knockout mice in mRNA and protein level (TGF-ß1: t = 1.945, P = 0.034; t = 1.902, P = 0.039; AP-1: t = 1.914, P = 0.038; t = 1.802, P = 0.041). CONCLUSIONS: p300 plays a crucial role in the pathogenesis of HSPN. p300 can down-regulate the expression of resistance genes (AP-1 and TGF-ß1) by binding with GRα to prevent further renal injury and glucocorticoid resistance. Therefore, p300 is a promising new target in glucocorticoid therapy in HSPN.


Asunto(s)
Dexametasona/uso terapéutico , Glucocorticoides/uso terapéutico , Vasculitis por IgA/tratamiento farmacológico , Vasculitis por IgA/metabolismo , Nefritis/tratamiento farmacológico , Nefritis/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Creatinina/sangre , Humanos , Vasculitis por IgA/sangre , Vasculitis por IgA/genética , Inmunoglobulina A/sangre , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis/sangre , Nefritis/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Factores de Transcripción p300-CBP/genética
20.
J Exp Biol ; 222(Pt 17)2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31371401

RESUMEN

Tissue repair is an adaptive and widespread metazoan response. It is characterised by different cellular mechanisms and complex signalling networks that involve numerous growth factors and cytokines. In higher animals, transforming growth factor-ß (TGF-ß) signalling plays a fundamental role in wound healing. In order to evaluate the involvement of TGF superfamily members in lower invertebrate tissue regeneration, sequences for putative TGF ligands and receptors were isolated from the transcriptome of the marine sponge Chondrosia reniformis We identified seven transcripts that coded for TGF superfamily ligands and three for TGF superfamily receptors. Phylogenetically, C. reniformis TGF ligands were not grouped into any TGF superfamily clades and thus presumably evolved independently, whereas the TGF receptors clustered in the Type I receptor group. We performed gene expression profiling of these transcripts in sponge regenerating tissue explants. Data showed that three ligands (TGF1, TGF3 and TGF6) were mainly expressed during early regeneration and seemed to be involved in stem cell maintenance, whereas two others (TGF4 and TGF5) were strongly upregulated during late regeneration and thus were considered pro-differentiating factors. The presence of a strong TGF inhibitor, SB431542, blocked the restoration of the exopinacoderm layer in the sponge explants, confirming the functional involvement of the TGF pathway in tissue regeneration in these early evolved animals.


Asunto(s)
Familia de Multigenes/fisiología , Poríferos/fisiología , Regeneración/genética , Factores de Crecimiento Transformadores/genética , Animales , Perfilación de la Expresión Génica , Factores de Crecimiento Transformadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA