Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Gastroenterol Hepatol ; 38(4): 634-647, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36807382

RESUMO

BACKGROUND: Genetic compensation response (GCR) is a mechanism that maintains the robustness of functional genes, which has been recently identified. Whether GCR exists in tumors and its effects on tumor progression remains unknown. METHODS: Whole exome sequencing was performed to identify premature termination codon (PTC) gene mutations in colorectal cancer (CRC) tissues. RNA sequencing, Cancer Cell Line Encyclopedia database analysis, and high-throughput output of homologous genes using the Ensemble genome database were performed to further identify homologous genes of target PTC gene mutations. RESULTS: Serine and arginine-rich splicing factor 3 (SRSF3) increased the invasion ability in CRC cells and could be the target gene of up-frameshift 3A (UPF3A). The deletion of the 660th base A in the coding sequence region of SRSF6 caused a frameshift mutation of serine at position 220 (s220fs), which contributed to a PTC UAA termination of translation in HCT116 cells. We further found that SRSF3 was the only homologue of SRSF6 with a frameshift mutation. The transfection of s220fs of SRSF6 into HCT116 cells led to upregulation of its corresponding oncogenic homologue gene SRSF3 expression to promote CRC metastasis. SRSF3 was highly expressed in CRC liver metastases and was positively correlated with UPF3A expression and contributed to poor prognosis. CONCLUSION: GCR may exist in CRC and exert effects on the progression of CRC. Targeted inhibition of UPF3A could reduce the GCR effects and suppress the expression of oncogenic homologue genes corresponding to PTC mutations, indicating a novel therapeutic strategy for treatment of CRC metastasis.


Assuntos
Neoplasias Colorretais , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina , Humanos , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica/genética , Células HCT116 , Fosfoproteínas , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina/genética , Fatores de Processamento de Serina-Arginina/metabolismo
2.
Insects ; 15(5)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38786869

RESUMO

Genetic mutations leading to premature termination codons are known to have detrimental effects. Using the Lepidoptera model insect, the silkworm (Bombyx mori), we explored the genetic compensatory response triggered by mutations with premature termination codons. Additionally, we delved into the molecular mechanisms associated with the nonsense-mediated mRNA degradation pathway. CRISPR/Cas9 technology was utilized to generate a homozygous bivoltine silkworm line BmTrpA1-/- with a premature termination. Transcript levels were assessed for the BmTrpA paralogs, BmPyrexia and BmPainless as well as for the essential factors Upf1, Upf2, and Upf3a involved in the nonsense-mediated mRNA degradation (NMD) pathway. Upf2 was specifically knocked down via RNA interference at the embryonic stage. The results comfirmed that the BmTrpA1 transcripts with a 2-base deletion generating a premature termination codon in the BmTrpA1-/- line. From day 6 of embryonic development, the mRNA levels of BmPyrexia, BmPainless, Upf1, and Upf2 were significantly elevated in the gene-edited line. Embryonic knockdown of Upf2 resulted in the suppression of the genetic compensation response in the mutant. As a result, the offspring silkworm eggs were able to hatch normally after 10 days of incubation, displaying a non-diapause phenotype. It was observed that a genetic compensation response does exist in BmTrpA1-/-B. mori. This study presents a novel discovery of the NMD-mediated genetic compensation response in B. mori. The findings offer new insights into understanding the genetic compensation response and exploring the gene functions in lepidopteran insects, such as silkworms.

3.
Cancer Manag Res ; 12: 4271-4281, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32606924

RESUMO

BACKGROUND: Nonsense-mediated mRNA decay (NMD) can degrade mRNAs with a premature termination codon (PTC), and undegraded mRNAs with PTC mutations can induce a genetic compensation response (GCR) by upregulating its compensatory genes. UPF3a refers to up-frame shift 3A (UPF3a) participating in NMD pathway and GCR. It inhibits the NMD pathway while it stimulates GCR. Notably, the role of UPF3a in cancer remains unclear. PURPOSE: The identification and discovery of prognostic markers for colorectal cancer (CRC) are of great clinical significance. The aim of this study was to investigate clinical significance of UPF3a expression in CRC. MATERIALS AND METHODS: UPF3a expression was examined in fresh CRC tissues and pared distant metastatic tissues using quantitative real-time PCR, Western blotting and immunohistochemistry staining. Tissue microarray immunohistochemical staining was used to study the relationship of UPF3a with clinicopathological features in 158 CRC patient samples collected from January 2008 to December 2012, and prognosis of CRC was analyzed. RESULTS: The expression of UPF3a was higher in metastatic tissues than that in primary sites. Moreover, high expression of UPF3a was significantly associated with TNM stage (p=0.009), liver metastasis and recurrence (p<0.001) in CRC patients. The Cancer Genome Atlas (TCGA) database showed the same trend. In CRC cells, knockdown of UPF3a led to a decline in the migration potential. Kaplan-Meier survival analysis revealed that high UPF3a expression, TNM stage were significantly associated (all P<0.01) with poor prognosis for patients. Furthermore, univariate and multivariate Cox analysis revealed that high UPF3a expression was independent risk factor for both overall survival and disease-free survival of CRC patients (all P<0.01). CONCLUSION: Results showed that high levels of UPF3a could lead to aggressiveness and poor CRC prognosis. Targeted UPF3a can act as a novel and effective gene therapy for CRC patients to make a better prognosis.

4.
Zebrafish ; 2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-32434440

RESUMO

The genetic compensation response (GCR), triggered by deleterious mutations but not by gene knockdown, has been proposed to explain many phenotypic discrepancies between gene-knockout and gene-knockdown models. GCRs have been observed in many model organisms from mice to Arabidopsis. Although the GCR is beneficial for organism survival, it impedes the exploration of gene function as many knockout mutants do not display discernible phenotypes due to the GCR. Uncovering how the mechanism of GCR operates is not only a fundamental goal in biology but also may provide a key solution in the unmasking of phenotypes in mutants displaying GCRs. Using zebrafish as the model, two recent studies have provided a molecular basis to explain this genetic paradox by demonstrating that the nonsense-mediated mRNA decay pathway is essential for nonsense mRNA to upregulate the expression of its homologous genes through an enhancement of histone H3 Lys4 trimethylation (H3K4me3) at the transcription start site regions of the compensatory genes. Here, we summarize the progress on the molecular mechanism of the GCR and make suggestions on how to overcome GCRs in the generation of genetic mutants.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA