Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Drug Dev Res ; 85(3): e22185, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38657094

RESUMEN

Cancer-associated fibroblasts (CAFs) play a pivotal role in cancer progression. This study aimed to explore the roles of CAFs-derived Fibroblast growth factor 9 (FGF9) and Neuro-oncological ventral antigen 1 (NOVA1) in triple negative breast cancer (TNBC) progression. MDA-MB-231 and BT-549 cells were cocultured with CAF conditioned-medium (CAF-CM) or normal fibroblasts conditioned-medium (NF-CM). MTT, EdU, colony formation, wound healing, transwell migration, and invasion assays were employed to determine cell proliferation, migration and invasion, respectively. Western blot and RT-qPCR were carried out to examine the protein and mRNA expression of FGF9 and NOVA1. Xenograft tumor experiments were conducted to evaluate the effects of CAFs, FGF9, and NOVA1 on tumor growth in vivo. Our results showed that CAFs significantly promoted the proliferation, invasion, and migration of TNBC cells. FGF9 and NOVA1 were significantly upregulated in TNBC CAFs, tissues and cells. CAF-CM also could increase the expression of FGF9 and NOVA1 in TNBC cells. Knockdown of FGF9 or NOVA1 could hamper cell proliferation, invasion, migration, and EMT of TNBC cells. Moreover, CAFs with FGF9/NOVA1 knockdown also could inhibit TNBC progression. Besides, CAFs significantly accelerated tumor growth in vivo, which was blocked by FGF9/NOVA1 knockdown in nude mice. In conclusion, our results indicated the tumor-promoting role of CAFs in TNBC progression. FGF9 and NOVA1 upregulation in CAFs induced cell proliferation, migration and invasion in vitro, and facilitated tumor growth in vivo in TNBC development.


Asunto(s)
Fibroblastos Asociados al Cáncer , Movimiento Celular , Proliferación Celular , Factor 9 de Crecimiento de Fibroblastos , Antígeno Ventral Neuro-Oncológico , Proteínas de Unión al ARN , Neoplasias de la Mama Triple Negativas , Animales , Femenino , Humanos , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Factor 9 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Proc Natl Acad Sci U S A ; 121(10): e2314695121, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38416679

RESUMEN

NOVA1 is a neuronal RNA-binding protein identified as the target antigen of a rare autoimmune disorder associated with cancer and neurological symptoms, termed paraneoplastic opsoclonus-myoclonus ataxia. Despite the strong association between NOVA1 and cancer, it has been unclear how NOVA1 function might contribute to cancer biology. In this study, we find that NOVA1 acts as an oncogenic factor in a GBM (glioblastoma multiforme) cell line established from a patient. Interestingly, NOVA1 and Argonaute (AGO) CLIP identified common 3' untranslated region (UTR) targets, which were down-regulated in NOVA1 knockdown GBM cells, indicating a transcriptome-wide intersection of NOVA1 and AGO-microRNA (miRNA) targets regulation. NOVA1 binding to 3'UTR targets stabilized transcripts including those encoding cholesterol homeostasis related proteins. Selective inhibition of NOVA1-RNA interactions with antisense oligonucleotides disrupted GBM cancer cell fitness. The precision of our GBM CLIP studies point to both mechanism and precise RNA sequence sites to selectively inhibit oncogenic NOVA1-RNA interactions. Taken together, we find that NOVA1 is commonly overexpressed in GBM, where it can antagonize AGO2-miRNA actions and consequently up-regulates cholesterol synthesis, promoting cell viability.


Asunto(s)
Glioblastoma , MicroARNs , Humanos , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , MicroARNs/genética , Homeostasis/genética , Colesterol , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Antígeno Ventral Neuro-Oncológico
3.
J Transl Med ; 21(1): 879, 2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-38049848

RESUMEN

BACKGROUND: Lung neuroendocrine neoplasms (LungNENs) comprise a heterogeneous group of tumors ranging from indolent lesions with good prognosis to highly aggressive cancers. Carcinoids are the rarest LungNENs, display low to intermediate malignancy and may be surgically managed, but show resistance to radiotherapy/chemotherapy in case of metastasis. Molecular profiling is providing new information to understand lung carcinoids, but its clinical value is still limited. Altered alternative splicing is emerging as a novel cancer hallmark unveiling a highly informative layer. METHODS: We primarily examined the status of the splicing machinery in lung carcinoids, by assessing the expression profile of the core spliceosome components and selected splicing factors in a cohort of 25 carcinoids using a microfluidic array. Results were validated in an external set of 51 samples. Dysregulation of splicing variants was further explored in silico in a separate set of 18 atypical carcinoids. Selected altered factors were tested by immunohistochemistry, their associations with clinical features were assessed and their putative functional roles were evaluated in vitro in two lung carcinoid-derived cell lines. RESULTS: The expression profile of the splicing machinery was profoundly dysregulated. Clustering and classification analyses highlighted five splicing factors: NOVA1, SRSF1, SRSF10, SRSF9 and PRPF8. Anatomopathological analysis showed protein differences in the presence of NOVA1, PRPF8 and SRSF10 in tumor versus non-tumor tissue. Expression levels of each of these factors were differentially related to distinct number and profiles of splicing events, and were associated to both common and disparate functional pathways. Accordingly, modulating the expression of NOVA1, PRPF8 and SRSF10 in vitro predictably influenced cell proliferation and colony formation, supporting their functional relevance and potential as actionable targets. CONCLUSIONS: These results provide primary evidence for dysregulation of the splicing machinery in lung carcinoids and suggest a plausible functional role and therapeutic targetability of NOVA1, PRPF8 and SRSF10.


Asunto(s)
Tumor Carcinoide , Neoplasias Pulmonares , Humanos , Tumor Carcinoide/genética , Tumor Carcinoide/metabolismo , Tumor Carcinoide/patología , Neoplasias Pulmonares/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Empalme Alternativo/genética , Factores de Empalme de ARN/genética , Biomarcadores/metabolismo , Biología , Pulmón/patología , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Antígeno Ventral Neuro-Oncológico
4.
Cells ; 12(20)2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37887320

RESUMEN

C9orf72 mutations are the most common form of familial amyotrophic lateral sclerosis (C9-ALS). It causes the production of proline-arginine dipeptide repeat proteins (PR-DPRs) in motor neurons (MNs), leading to the molecular pathology characteristic of ALS. UNC13A is critical for maintaining the synaptic function of MNs. Most ALS patients have nuclear deletion of the splicing repressor TDP-43 in MNs, which causes inclusion of the cryptic exon (CE) of UNC13A mRNA, resulting in nonsense-mediated mRNA decay and reduced protein expression. Therefore, in this study, we explored the role of PR-DPR in CE inclusion of UNC13A mRNA. Our results showed that PR-DPR (PR50) induced CE inclusion and decreased the protein expression of UNC13A in human neuronal cell lines. We also identified an interaction between the RNA-binding protein NOVA1 and PR50 by yeast two-hybrid screening. NOVA1 expression is known to be reduced in patients with ALS. We found that knockdown of NOVA1 enhanced CE inclusion of UNC13A mRNA. Furthermore, the naturally occurring triterpene betulin can inhibit the interaction between NOVA1 and PR50, thus preventing CE inclusion of UNC13A mRNA and protein reduction in human neuronal cell lines. This study linked PR-DPR with CE inclusion of UNC13A mRNA and developed candidate therapeutic strategies for C9-ALS using betulin.


Asunto(s)
Esclerosis Amiotrófica Lateral , Humanos , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Arginina/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Dipéptidos/metabolismo , Neuronas Motoras/patología , Antígeno Ventral Neuro-Oncológico , Prolina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
5.
Elife ; 122023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37347149

RESUMEN

Somatostatin interneurons are the earliest born population of cortical inhibitory cells. They are crucial to support normal brain development and function; however, the mechanisms underlying their integration into nascent cortical circuitry are not well understood. In this study, we begin by demonstrating that the maturation of somatostatin interneurons in mouse somatosensory cortex is activity dependent. We then investigated the relationship between activity, alternative splicing, and synapse formation within this population. Specifically, we discovered that the Nova family of RNA-binding proteins are activity-dependent and are essential for the maturation of somatostatin interneurons, as well as their afferent and efferent connectivity. Within this population, Nova2 preferentially mediates the alternative splicing of genes required for axonal formation and synaptic function independently from its effect on gene expression. Hence, our work demonstrates that the Nova family of proteins through alternative splicing are centrally involved in coupling developmental neuronal activity to cortical circuit formation.


Asunto(s)
Empalme Alternativo , Interneuronas , Ratones , Animales , Interneuronas/fisiología , Neuronas/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Somatostatina/metabolismo
6.
Cell Rep ; 42(2): 112050, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36716149

RESUMEN

We describe a patient haploinsufficient for the neuronal RNA binding protein NOVA1 who developed a behavioral motor hyperactivity disorder, suggesting a role of NOVA1 in postnatal motor inhibition. To investigate Nova1's action in adult Gad2+ inhibitory neurons, we generated a conditional Nova1-null mouse (Nova1-cKOGad2-cre). Strikingly, the phenotypes of these mice show many similarities to the NOVA1 haploinsufficient patient and identify a function of Nova1 in the hypothalamus. Molecularly, Nova1 loss in Gad2-positive neurons alters downstream expression of Impact mRNA, along with a subset of RNAs encoding electron transport chain-related factors and ribosomal proteins. NOVA1 stabilizes Impact mRNA by binding its 3' UTR, antagonizing the actions of miR-138 and miR-124. Together, these studies demonstrate actions of NOVA1 in adult hypothalamic neurons, mechanisms by which it functions in translation and metabolism, including through direct binding to Impact mRNA, and illuminate its role in human neurologic disease.


Asunto(s)
MicroARNs , Antígeno Ventral Neuro-Oncológico , Proteínas de Unión al ARN , Animales , Humanos , Ratones , Hipotálamo/metabolismo , MicroARNs/metabolismo , Antígeno Ventral Neuro-Oncológico/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
7.
BMC Cancer ; 22(1): 1091, 2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36284263

RESUMEN

BACKGROUND: Neuro-oncological ventral antigen 1 (NOVA1) is a neuron-specific RNA-binding protein which regulates alternative splicing in the developing nervous system. Recent research has found that NOVA1 plays a significant role in carcinogenesis. In this paper, we examine the role of NOVA1 in non-small cell lung cancer (NSCLC) and its underlying molecular mechanisms. METHODS: The expression of NOVA1 in NSCLC was detected by immunohistochemistry and correlations between NOVA1 expression and clinicopathological factors were analyzed by chi-square tests. Kaplan-Meier survival analysis and the Cox regression model were used to evaluate the predictive effect of prognostic factors. Western blotting, Cell Counting Kit-8, colony formation, apoptosis, migration and invasion assays were used to detect the effects of silencing (si)NOVA1 RNA on Wnt/ß-catenin signaling and biological behavior in NSCLC cell lines. RESULTS: Our study showed that expression of NOVA1 was up-regulated and significantly correlated with poor differentiation (p = 0.020), advanced TNM stage (P = 0.001), T stage (P = 0.001) and lymph node metastasis (P = 0.000) as well as the expression of ß-catenin (P = 0.012) in NSCLC. The down-regulation of NSCLC by siRNA significantly inhibited proliferation, migration and invasion and promoted apoptosis in NSCLC cells. Expression of Wnt signaling molecules, including ß-catenin, activated ß-catenin, cyclin D1, matrix metalloproteinase (MMP)-2 and MMP-7, was also significantly reduced by siNOVA1. The inhibition of Wnt/ß-catenin signaling in A549 and H1299 cells by siNOVA1 was reversed after treatment with a ß-catenin expression plasmid. CONCLUSION: The present study suggests that NOVA1 may serve as a potential prognosis biomarker in NSCLC. High NOVA1 expression was associated with poor survival rate. Finally, in vitro experiments verified that NOVA1 promotes NSCLC cell proliferation and invasion by regulating Wnt/ß-catenin signaling.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Vía de Señalización Wnt , Ciclina D1/genética , Metaloproteinasa 7 de la Matriz/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Neoplasias Pulmonares/patología , ARN Interferente Pequeño/farmacología , Antígeno Ventral Neuro-Oncológico , Proliferación Celular/genética , Pronóstico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Biomarcadores , Movimiento Celular/genética , Línea Celular Tumoral
8.
Nutrients ; 14(18)2022 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-36145242

RESUMEN

The loss of functional pancreatic ß-cell mass is an important hallmark of both type 1 and type 2 diabetes. The RNA-binding protein NOVA1 is expressed in human and rodent pancreatic ß-cells. Previous in vitro studies indicated that NOVA1 is necessary for glucose-stimulated insulin secretion and its deficiency-enhanced cytokine-induced apoptosis. Moreover, Bim, a proapoptotic protein, is differentially spliced and potentiates apoptosis in NOVA1-deficient ß-cells in culture. We generated two novel mouse models by Cre-Lox technology lacking Nova1 (ßNova1-/-) or Bim (ßBim-/-) in ß-cells. To test the impact of Nova1 or Bim deletion on ß-cell function, mice were subjected to multiple low-dose streptozotocin (MLD-STZ)-induced diabetes or high-fat diet-induced insulin resistance. ß-cell-specific Nova1 or Bim deficiency failed to affect diabetes development in response to MLD-STZ-induced ß-cell dysfunction and death evidenced by unaltered blood glucose levels and pancreatic insulin content. In addition, body composition, glucose and insulin tolerance test, and pancreatic insulin content were indistinguishable between control and ßNova1-/- or ßBim-/- mice on a high fat diet. Thus, Nova1 or Bim deletion in ß-cells does not impact on glucose homeostasis or diabetes development in mice. Together, these data argue against an in vivo role for the Nova1-Bim axis in ß-cells.


Asunto(s)
Proteína 11 Similar a Bcl2/metabolismo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Glucemia/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Glucosa/metabolismo , Humanos , Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Antígeno Ventral Neuro-Oncológico , Obesidad/etiología , Obesidad/metabolismo , Proteínas de Unión al ARN/metabolismo , Estreptozocina
9.
J Cardiovasc Transl Res ; 15(5): 1176-1191, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35377129

RESUMEN

Angina pectoris is the most common manifestation of coronary heart disease, causing suffering in patients. Electroacupuncture at PC6 can effectively alleviate angina by regulating the expression of genes, whether the alternative splicing (AS) of genes is affected by acupuncture is still unknown. We established a rat model of myocardial ischemia-reperfusion by coronary artery ligation and confirmed electroacupuncture alleviated the abnormal discharge caused by angina pectoris measured in EMG electromyograms. Analysis of the GSE61840 dataset established that AS events were altered after I/R and regulated by electroacupuncture. I/R decreased the expression of splicing factor Nova1 while electroacupuncture rescued it. Further experiments in dorsal root ganglion cells showed Nova1 regulated the AS of the GABRG2, specifically on its exon 9 where an important phosphorylation site is present. In vivo, results also showed that electroacupuncture can restore AS of GABRG2. Our results proved that electroacupuncture alleviates angina results by regulating alternative splicing.


Asunto(s)
Electroacupuntura , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , Animales , Ratas , Puntos de Acupuntura , Empalme Alternativo , Angina de Pecho , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/prevención & control , Receptores de Neurotransmisores , Proteínas de Unión al ARN/genética
10.
Bioengineered ; 13(4): 8982-8993, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35348441

RESUMEN

MiRNAs are a class of small non-coding RNAs (ncRNAs) responsible for post-transcriptional regulation of target genes. Accumulating evidence indicates that miRNAs are implicated in the progression of cardiac hypertrophy. Therefore, understanding the molecular mechanisms how these miRNAs regulate cardiac hypertrophy is useful for diagnosis and monitoring of disease progression. In this study, to investigate the effect of miR-27a-3p, we established an in vitro cardiac hypertrophy model by treating H9c2 cardiomyocytes with angiotensin II (Ang II) and an in vivo model through the chronic infusion of Ang II into mice. As revealed by our experimental results, miR-27a-3p expression was significantly increased in clinical samples, animal and cell models of cardiac hypertrophy. Inhibiting miR-27a-3p mitigated cardiac hypertrophy phenotype induced by Ang II. Additionally, our work identified NOVA1 (neuro-oncological ventral antigen 1) as a downstream target of miR-27a-3p. miR-27a-3p overexpression reduced NOVA1 protein level and mRNA expression. Consistently, NOVA1 silencing promoted cardiac hypertrophy phenotype induced by Ang II. In summary, these results suggest that the upregulation of miR-27a-3p may serve as a diagnostic factor for cardiac hypertrophy, and miR-27a-3p upregulation promotes cardiac hypertrophy by targeting NOVA1.


Asunto(s)
Cardiomegalia , MicroARNs , Antígeno Ventral Neuro-Oncológico , Angiotensina II , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Ratones , MicroARNs/genética , Miocitos Cardíacos/metabolismo
11.
Front Cell Dev Biol ; 9: 673873, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34692669

RESUMEN

Background: Gene expression and alternative splicing (AS) can promote cancer development via complex mechanisms. We aimed to identify and verify the hub AS events and splicing factors associated with the progression of colorectal cancer (CRC). Methods: RNA-Seq data, clinical data, and AS events of 590 CRC samples were obtained from the TCGA and TCGASpliceSeq databases. Cox univariable and multivariable analyses, KEGG, and GO pathway analyses were performed to identify hub AS events and splicing factor/spliceosome genes, which were further validated in five CRCs. Results: In this study, we first compared differentially expressed genes and gene AS events between normal and tumor tissues. Differentially expressed genes were different from genes with differentially expressed AS events. Prognostic analysis and co-expression network analysis of gene expression and gene AS events were conducted to screen five hub gene AS events involved in CRC progression: EPB41L2, CELF2, TMEM130, VCL, and SORBS2. Using qRT-PCR, we also verified that the gene AS events SORBS2 were downregulated in tumor tissue, and gene AS events EPB41L2, CELF2, TMEM130, and VCL were upregulated in tumor tissue. The genes whose mRNA levels were significantly related to the five hub gene AS events were significantly enriched in the GO term of cell division and Notch signaling pathway. Further coexpression of gene AS events and alternative splicing factor genes revealed NOVA1 as a crucial factor regulating the hub gene AS event expression in CRC. Through in vitro experiments, we found that NOVA1 inhibited gene AS event SORBS2, which induced the migration of CRC cells via the Notch pathway. Conclusion: Integrated analysis of gene expression and gene AS events and further experiments revealed that NOVA1-mediated SORBS2 promoted the migration of CRC, indicating its potential as a therapeutic target.

12.
Mol Immunol ; 138: 31-37, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34332183

RESUMEN

NOVA1 (neuro-oncological ventral antigen 1) is a neuron specific RNA binding protein, belonging to the Nova family, which plays an important role in various diseases. However, the role of NOVA1 in childhood asthma remains unclear. This study was aimed to investigate the role of NOVA1 in TGF-ß1-induced ASMCs proliferation and migration as well as the potential mechanisms. In our study, the NOVA1 expression was significantly increased in asthmatic tissues and TGF-ß1-induced ASMCs. Inhibition of NOVA1 significantly inhibited TGF-ß1-induced ASMCs cell proliferation and migration, and alleviates TGF-ß1-induced inflammation. NOVA1 positively regulated the PTBP1 expression and si-NOVA1 inhibited the activation of PTEN/AKT signal pathway. Importantly, the overexpression of PTBP1 partially reversed the effect of NOVA1 on cell viability, migration, inflammation and the activation of PTEN/AKT signal pathway. Generally, our study demonstrated that si-NOVA1 inhibited TGF-ß1-induced inflammation and migration in ASMCs through PTBP1/PTEN/AKT pathway. Therefore, inhibition of NOVA1 may be useful for the prevention or treatment of asthma airway remodeling.


Asunto(s)
Asma/patología , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/fisiología , Adolescente , Remodelación de las Vías Aéreas (Respiratorias) , Asma/metabolismo , Asma/fisiopatología , Movimiento Celular/fisiología , Niño , Preescolar , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Miocitos del Músculo Liso/patología , Antígeno Ventral Neuro-Oncológico , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
13.
Genes (Basel) ; 12(3)2021 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-33799408

RESUMEN

The movement of abnormal vascular smooth muscle cells (VSMCs) contributes to intimal hyperplasia in vein graft disease. Circular RNAs (circRNAs) are single stranded RNAs with 3' and 5' ends covalently joined together. They have been shown to regulate cell function in many diseases. NOVA1 is considered to be a brain-specific splicing factor that plays an important role in the nervous system and cancer. The role of NOVA1 in VSMCs remains unclear. In the present study, transcriptome sequencing was used to identify differentially expressed circRNAs in the rat vein graft model. A novel circRNA, circUVRAG, was decreased in the grafted vein and stably located in the cytoplasm. Knockdown of circUVRAG suppressed VSMC adhesion and migration. In addition, we demonstrated that the alternative splicing factor NOVA1 co-located with UVRAG pre-mRNA in the nucleus and modulated the production of circUVRAG. These new discoveries may serve as a potential means to treat intimal hyperplasia after vein grafts.


Asunto(s)
Empalme Alternativo , Movimiento Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Circular/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Adhesión Celular , Masculino , Antígeno Ventral Neuro-Oncológico , ARN Circular/genética , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley
14.
Mol Cell Biochem ; 476(1): 279-292, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32965597

RESUMEN

Drug resistance is one of the major challenges for cancer therapies. In recent years, research on disease-related molecular signaling pathways has become the key ways to understand and overcome obstacles. Dysregulation of MALAT1 could regulate doxorubicin resistance of hepatocellular carcinoma (HCC), but how MALAT1 involving in managing doxorubicin resistance remains unclear yet. We aimed to elucidate the specific molecular mechanism of MALAT1 with doxorubicin resistance in HCC cells. Quantitative real-time polymerase chain reaction (qRT-PCR) was engaged to detect the expression levels of MALAT1, miR-3129-5p and Nova1 mRNA; MTT, western blot, flow cytometry and luciferase reporter assays were executed to identify the influence of MALAT1 on doxorubicin resistance of HCC cells. Xenograft tumor model was created to confirm the biological function of MALAT1 in doxorubicin resistance of HCC cells in vivo. MALAT1 and Nova1 were upregulated, while miR-3129-5p expression was decreased in doxorubicin-resistant HCC tissues and cells. Knockdown of MALAT1 regulated doxorubicin resistance of HCC cells through inhibiting cell proliferation, migration, invasion and promoting apoptosis, but antisense miR-3129-5p released the functional effect of MALAT1 knockdown. Nova1, as a target gene of miR-3129-5p, reversed the results of miR-3129-5p expression and enhanced doxorubicin resistance of HCC cells. Xenograft tumor model suggested that dysregulation of MALAT1 regulated tumor growth and Nova1 to mediate doxorubicin resistance of HCC cells by as a sponge for miR-3129-5p in vivo. Elevation of LncRNA MALAT1 mediated doxorubicin resistance and the progression of HCC via a MALAT1/miR-3129-5p/Nova1 axis. This study would be expected to enrich the understanding of doxorubicin resistance of HCC and provide new ideas for HCC treatment strategies.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Antígeno Ventral Neuro-Oncológico , Transducción de Señal
15.
Kaohsiung J Med Sci ; 37(4): 286-293, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33340248

RESUMEN

MicroRNAs (miRNAs) are dysregulated in many tumors and have been found to play crucial roles in cancer biology. Retinoblastoma is a rare tumor that develops rapidly from a malignant tumor of immature cells in the retina known as photoreceptor progenitors. Our study aimed to explore the role of miR-146a in the pathology of retinoblastoma. Potential target gene of miR-146a was predicted by Targetscan. Reverse transcription quantitative polymerase chain reaction (RT-PCR) showed that miR-146a was downregulated and ventral nerve tumor antigen 1 (Neuro - oncological ventral antigen 1, NOVA1) was upregulated in retinoblastoma. Luciferase assay confirmed that miR-146a directly target NOVA1. MiR-146a knockdown and overexpression experiments were performed and found that miR-146a could regulate the expression of NOVA1. The miR-146a knockdown and overexpression experiments were conducted to investigate the biological function of miR-146a. MiR-146a was found inhibited the viability, proliferation and invasion of retinoblastoma cell by MTT, EdU, and transwell assays. Flow cytometry was performed for the apoptosis analysis and miR-146a increased the apoptosis of retinoblastoma cell was found. Above phenomenon can be rescued by overexpression of NOVA1. In conclusion, these results suggest that miR-146a acts as a tumor suppressor and can act as a potential therapeutic target for retinoblastoma in the future.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteínas de Unión al ARN/genética , Neoplasias de la Retina/genética , Retinoblastoma/genética , Apoptosis/genética , Emparejamiento Base , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Preescolar , Femenino , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , MicroARNs/metabolismo , Antígeno Ventral Neuro-Oncológico , Proteínas de Unión al ARN/metabolismo , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/metabolismo , Retinoblastoma/patología , Transducción de Señal
16.
Aging (Albany NY) ; 12(13): 12841-12849, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32589614

RESUMEN

Diabetic wounds increase morbidity and decrease quality of life in patients with type 2 diabetes. Serum miR-27-3p levels are reportedly elevated in type 2 diabetic patients. In the present study, we explored the role of miR-27-3p during wound healing. We found that miR-27-3p is overexpressed in cutaneous fibroblasts of diabetic patients and mice. miR-27-3p knockdown enhanced the proliferation and migration of fibroblasts, while suppressing the incidence of fibroblast apoptosis. Overexpressing miR-27-3p in fibroblasts had the opposite effects. We also identified neuro-oncological ventral antigen 1 (NOVA1) as a target of miR-27-3p in fibroblasts. Knocking down NOVA1 using targeted siRNA mimicked the effects of miR-27-3p overexpression in fibroblasts. Administration of miR-27-3p to the area around wounds inflicted in mice delayed healing of those wounds. This suggests that miR-27-3p suppresses fibroblast function by targeting NOVA1, which results in the slowing of wound healing. These findings may offer a new approach to the treatment of diabetic wound healing.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , MicroARNs , Proteínas de Unión al ARN , Cicatrización de Heridas/fisiología , Anciano , Animales , Apoptosis , Supervivencia Celular , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Antígeno Ventral Neuro-Oncológico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
17.
Int J Mol Sci ; 21(11)2020 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-32486302

RESUMEN

NOVA1 and NOVA2, the two members of the NOVA family of alternative splicing factors, bind YCAY clusters of pre-mRNAs and assemble spliceosomes to induce the maintenance/removal of introns and exons, thus governing the development of mRNAs. Members of other splicing families operate analogously. Activity of NOVAs accounts for up to 700 alternative splicing events per cell, taking place both in the nucleus (co-transcription of mRNAs) and in the cytoplasm. Brain neurons express high levels of NOVAs, with NOVA1 predominant in cerebellum and spinal cord, NOVA2 in the cortex. Among brain physiological processes NOVAs play critical roles in axon pathfinding and spreading, structure and function of synapses, as well as the regulation of surface receptors and voltage-gated channels. In pathology, NOVAs contribute to neurodegenerative diseases and epilepsy. In vessel endothelial cells, NOVA2 is essential for angiogenesis, while in adipocytes, NOVA1 contributes to regulation of thermogenesis and obesity. In many cancers NOVA1 and also NOVA2, by interacting with specific miRNAs and by additional mechanisms, activate oncogenic roles promoting cell proliferation, colony formation, migration, and invasion. In conclusion, NOVAs regulate cell functions of physiological and pathological nature. Single cell identification and distinction, and new therapies addressed to NOVA targets might be developed in the near future.


Asunto(s)
Empalme Alternativo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Adipocitos/citología , Mapeo Encefálico , Movimiento Celular , Proliferación Celular , Células Endoteliales/metabolismo , Humanos , Invasividad Neoplásica , Neoplasias/metabolismo , Neoplasias/terapia , Neovascularización Patológica , Proteínas del Tejido Nervioso/genética , Antígeno Ventral Neuro-Oncológico , Neuronas/metabolismo , Obesidad/metabolismo , Obesidad/terapia , Proteínas de Unión al ARN/genética , Empalmosomas/metabolismo
18.
Transl Cancer Res ; 9(2): 1144-1150, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35117458

RESUMEN

BACKGROUND: To investigate the expression of neuro-oncological ventral antigen-1 (Nova1) in small-cell lung cancer (SCLC) and its value in clinical pathological diagnosis of SCLCs. METHODS: Nova1 expression in the SCLC cell line H446, lung adenocarcinoma cell line A549, squamous cell carcinoma (SCC) cell line H226 and bronchial epithelial cell line BEAS-2B were examined by RT-PCR. It's expression in paraffin specimens of 100 SCLC cases, 15 specimens of lung adenocarcinoma cases and 15 specimens of lung SCC cases, as well as adjacent normal tissue was determined by immunohistochemistry. The positive rate of Nova1 in SCLC tissue was compared with those of traditional neuroendocrine markers CD56, Syn and CgA. RESULTS: Nova1 expression in SCLC cell line H446 was significantly higher than that of bronchial epithelial cell line BEAS-2B and lung adenocarcinoma cell line A549 and SCC cell line H226. The positive rate of Nova1 in SCLC tissue was similar to that of CD56 but higher than those of Syn and CgA. Nova1 was neither expressed in the cancer tissue nor in the adjacent normal tissue of the 15 cases of SCC and the 15 cases of adenocarcinoma. In all six cases of combined SCLC, Nova1 was expressed in the SCLC component, whereas markers of SCC or adenocarcinoma were absent. CONCLUSIONS: Nova1 can be used as a novel neuroendocrine marker for pathological diagnosis of SCLC in clinical practice with high sensitivity and specificity, in addition to its role as an independent prognostic marker of overall survival and tumour recurrence, as suggested by previous studies.

19.
Transl Cancer Res ; 9(7): 4373-4382, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35117803

RESUMEN

BACKGROUND: Small cell lung cancer (SCLC) is a highly aggressive lung malignancy which is characteristic of rapid tumor proliferation, metastasis as well as paraneoplastic neurological syndromes (PNS). Recent studies have shown that neuro-oncological ventral antigen-1 (NOVA1) plays a crucial role in the progression of various tumors. However, its effect on SCLC is still exclusive. This study was aimed to explore NOVA1 expression in tumor tissues of SCLC patients as well as its correlation with clinicopathological characteristics and cancer-specific overall survivals. METHODS: In this study, the patients pathologically diagnosed with new primary SCLC were enrolled. Firstly, we compared NOVA1 expression in SCLC tissues and adjacent normal tissues by immunohistochemistry. We further investigated the association of NOVA1 expression with clinicopathological markers (especially the categories of PNS) and survival time in SCLC patients. RESULTS: Our finding exhibited that NOVA1 was remarkably expressed in SCLC tumorous tissues compared with adjacent normal lung tissues (P<0.001). Additionally, NOVA1 expression was closely associated with clinicopathological characteristics in SCLC patients in terms of tumor staging(χ2=15.833; P<0.001), lymph node metastasis (χ2=9.624; P=0.002), brain metastasis (χ2=9.624; P=0.002) and PNS (χ2=5.004; P=0.024). With the COX proportional hazard regression model, we detected that high expression of NOVA1 (HR =0.445; 95% CI: 0.213-0.934; P=0.032) was an independent factor for shorter survival time. CONCLUSIONS: High expression of NOVA1 is closely associated with aggressive clinicopathological characteristics including PNS as well as poor survival in SCLC patients. NOVA1 can be served as a promising predictive factor for prognosis in SCLC.

20.
Surg Oncol ; 31: 67-74, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31541909

RESUMEN

The molecular mechanisms governing the metastasis of colorectal cancer (CRC) are incompletely understood. In the present study, we found NOVA1 to be expressed at higher levels in CRC cell lines and tissue samples, and this upregulation was positively correlated with TNM stage (p = 0.034), poor differentiation (p = 0.001), and lymph node metastasis (p = 0.008). Both overall survival (OS) and relapse-free survival (RFS) were both significantly decreased in patients with high NOVA1 expression relative to those with low expression. Through a multivariate analysis, we determined that NOVA1 independently predicted poor outcomes in those with CRC. In further functional studies, we found that NOVA1 expression controlled the proliferation and invasive characteristics of CRC cells via a mechanism wherein NOVA1 bound and stabilized the IL6 mRNA, enhancing IL-6/JAK2/STAT3 signaling to in turn upregulate matrix metalloproteinases (MMPs) 2, 7, and 9. NOVA1 therefore plays key functional roles in regulating CRC progression, and our results further indicate that it serve as a valuable prognostic biomarker and potentially a target for therapeutic treatment in individuals with CRC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Interleucina-6/genética , Janus Quinasa 2/metabolismo , Neoplasias Pulmonares/secundario , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Janus Quinasa 2/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Antígeno Ventral Neuro-Oncológico , Pronóstico , Estabilidad del ARN , Proteínas de Unión al ARN/genética , Factor de Transcripción STAT3/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA