Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(2): 76, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36725842

RESUMO

Although programmed death-ligand 1 (PD-L1) inhibitors have achieved some therapeutic success in breast cancer, their efficacy is limited by low therapeutic response rates, which is closely related to the immune escape of breast cancer cells. Tissue differentiation inducing non-protein coding RNA (TINCR), a long non-coding RNA, as an oncogenic gene associated with the progression of various malignant tumors, including breast cancer; however, the role of TINCR in tumor immunity, especially in breast cancer, remains unclear. We confirmed that TINCR upregulated PD-L1 expression in vivo and in vitro, and promoted the progression of breast cancer. Next, we revealed that TINCR knockdown can significantly improve the therapeutic effect of PD-L1 inhibitors in breast cancer in vivo. Mechanistically, TINCR recruits DNMT1 to promote the methylation of miR-199a-5p loci and inhibit its transcription. Furthermore, in the cytoplasm, TINCR potentially acts as a molecular sponge of miR-199a-5p and upregulates the stability of USP20 mRNA through a competing endogenous RNA (ceRNA) regulatory mechanism, thus promoting PD-L1 expression by decreasing its ubiquitination level. IFN-γ stimulation activates STAT1 by phosphorylation, which migrates into the nucleus to promote TINCR transcription. This is the first study to describe the regulatory role of TINCR in breast cancer tumor immunity, broadening the current paradigm of the functional diversity of TINCR in tumor biology. In addition, our study provides new research directions and potential therapeutic targets for PD-L1 inhibitors in breast cancer.


Assuntos
Neoplasias da Mama , MicroRNAs , RNA Longo não Codificante , Feminino , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Inibidores de Checkpoint Imunológico , Imunoterapia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Ubiquitina Tiolesterase/metabolismo
3.
Arch Med Sci ; 18(2): 422-431, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35316902

RESUMO

Introduction: The present study aims to clarify the advantages and disadvantages of elite biopsy, to provide a reference for selecting the puncture method. Material and methods: A total of 802 patients with a BI-RADS grade ≥ 4, as evaluated by the molybdenum target, and measurable lesions revealed by colour Doppler ultrasound, who were admitted at our department from January 2017 to January 2018, were enrolled in the present study. These patients were randomly divided into three groups: elite, Mammotome and core needle biopsy groups. The pathological underestimation rate, diagnostic accordance rate, haematoma incidence rate, and costs of these three biopsy methods were compared. Results: The difference in diagnostic accordance rates between the elite biopsy group and core needle biopsy group was statistically significant (98.9% vs. 94.7%, p = 0.003), as well as between the Mammotome biopsy group and core needle biopsy group (99.6% vs. 94.7%, p < 0.001). The difference in pathological underestimation rates between the elite biopsy group and core needle biopsy group was statistically significant (7.2% vs. 37.3%, p < 0.001), as well as between the Mammotome biopsy group and core needle biopsy group (1.6% vs. 7.2%, p < 0.001). The difference between the Mammotome biopsy group and elite biopsy group was not statistically significant. The incidence of haematoma in the Mommotome, elite, and core needle groups was 15.9%, 13%, and 21.7%, respectively (13% vs. 21.7%, p = 0.021). Conclusions: Elite biopsy has a low rate of pathological underestimation and low incidence of haematoma, can improve the breast conserving rate, and has an affordable cost. As a biopsy method with high accuracy, safety, and economy, elite biopsy can be widely used in clinics.

4.
Cell Death Dis ; 13(2): 177, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210409

RESUMO

While cells activate a multifaceted DNA damage response to remove transcription-blocking DNA lesions, mechanisms to regulate genome-wide reduction of RNA synthesis and the paradoxical continuous loading of RNAP II at initiation sites are still poorly understood. Uncovering how dramatic changes to the transcriptional program contribute to TC-NER (transcription-coupled nucleotide excision repair) is important in DNA repair research. However, the functional significance of transcriptome dynamics and the mechanisms of chromatin attachment for thousands of unstudied human lncRNAs remain unclear. To address these questions, we examined UV-induced gene expression regulation in human fibroblasts by performing RNA-seq with fractionated chromatin-associated and cytoplasmic transcripts. This approach allowed us to separate the synthesis of nascent transcripts from the accumulation of mature RNAs. In addition to documenting the subcellular locations of coding transcripts, our results also provide a high-resolution view of the transcription activities of noncoding RNAs in response to cellular stress. At the same time, the data showed that vast majority of genes exhibit large changes in chromatin-associated nascent transcripts without corresponding changes in cytoplasmic mRNA levels. Distinct from protein-coding genes that transcripts with shorter length prefer to be recovered first, repression of lncRNA transcription after UV exposure is inactivated first on noncoding transcripts with longer length. This work provides an updated framework for cellular RNA organization in response to stress and may provide useful information in understanding how cells respond to transcription-blocking DNA damage.


Assuntos
RNA Longo não Codificante , Transcriptoma , Cromatina/genética , Cromatina/metabolismo , Reparo do DNA/genética , Fibroblastos/metabolismo , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transcrição Gênica , Transcriptoma/genética
5.
Cell Biosci ; 12(1): 22, 2022 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-35216622

RESUMO

Breast cancer is the most prevalent malignant tumor and a leading cause of mortality among females worldwide. The tumorigenesis and progression of breast cancer involve complex pathophysiological processes, which may be mediated by post-translational modifications (PTMs) of proteins, stimulated by various genes and signaling pathways. Studies into PTMs have long been dominated by the investigation of protein phosphorylation and histone epigenetic modifications. However, with great advances in proteomic techniques, several other PTMs, such as acetylation, glycosylation, sumoylation, methylation, ubiquitination, citrullination, and palmitoylation have been confirmed in breast cancer. Nevertheless, the mechanisms, effects, and inhibitors of these unconventional PTMs (particularly, the non-histone modifications other than phosphorylation) received comparatively little attention. Therefore, in this review, we illustrate the functions of these PTMs and highlight their impact on the oncogenesis and progression of breast cancer. Identification of novel potential therapeutic drugs targeting PTMs and development of biological markers for the detection of breast cancer would be significantly valuable for the efficient selection of therapeutic regimens and prediction of disease prognosis in patients with breast cancer.

6.
Cell Biosci ; 11(1): 169, 2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34465388

RESUMO

BACKGROUND: Long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) plays a positive role in the progression of human malignant tumors. However, the molecular mechanism of SNHG1 remains elusive in breast cancer. RESULTS: LncRNA SNHG1 was upregulated and had a positive relationship with poor prognosis according to bioinformatics analysis in pan-cancer including breast cancer. Silencing SNHG1 inhibited tumorigenesis in breast cancer both in vitro and in vivo. Mechanistically, SNHG1 functioned as a competing endogenous RNA (ceRNA) to promote TERT expression by sponging miR-18b-5p in breast cancer. miR-18b-5p acted as a tumor repressor in breast cancer. Moreover, the combination of SNHG1 knockdown and TERT inhibitor administration showed a synergistic inhibitory effect on breast cancer growth in vivo. Finally, E2F1 as a transcription factor, binding to SNHG1 promoter and enhanced SNHG1 transcription in breast cancer. CONCLUSIONS: Our results provide a comprehensive understanding of the oncogenic mechanism of lncRNA SNHG1 in breast cancer. Importantly, we identified a novel E2F1-SNHG1-miR-18b-5p-TERT axis, which may be a potential therapeutic target for breast cancer. Our results also provided a potential treatment for breast cancer when knockdown SNHG1 and TERT inhibitor administration simultaneously.

7.
DNA Repair (Amst) ; 106: 103172, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34298489

RESUMO

The transcription-related DNA damage response was visualized on a genome-wide scale with great spatial and temporal resolution. Upon UV irradiation, a small proportion of mature RNA transcripts undergo changes, with significant activation of DNA repair factors. Notably, an increase of chromatin accessibility is observed at the immediate early recovery phase and serves as binding sites for selective stage-specific transcription factors. Whole genome analysis of DNA methylation (5mC) delineates pervasive dynamics during DNA repair process, and hypomethylation at gene bodies and 3'UTR is accompanied by induction of DNA damage response genes. Furthermore, temporal-specific m6A RNA methylation has been defined and appears to affect DNA repair by modulation of translation. These findings provide a resource for identifying players required for transcription-coupled nucleotide excision repair and reveal insights into the epigenetic regulation of the transcriptional programs in response to genotoxic stress.


Assuntos
Metilação de DNA , Reparo do DNA , Epigênese Genética , Fibroblastos/efeitos da radiação , RNA/metabolismo , Transcriptoma , Raios Ultravioleta , Adenosina/análogos & derivados , Linhagem Celular , DNA/metabolismo , Dano ao DNA , Epigenômica , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Metilação , Processamento Pós-Transcricional do RNA , Transcrição Gênica
8.
Cell Death Dis ; 12(1): 83, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446634

RESUMO

The long noncoding RNA (lncRNA) TINCR has recently been found to be associated with the progression of human malignancies, but the molecular mechanism of TINCR action remains elusive, particularly in breast cancer. The oncogenic role of TINCR was examined in vitro and in vivo in breast cancer. Next, the interaction between TINCR, DNMT1, and miR-503-5p methylation was explored. Moreover, the mechanism by which TINCR enhances EGFR expression and downstream signaling via an RNA-RNA interaction was comprehensively investigated. Furthermore, upstream transcriptional regulation of TINCR expression by STAT3 was examined by performing chromatin immunoprecipitation. Finally, feedback signaling in the STAT3-TINCR-EGFR downstream cascade was also investigated. TINCR is upregulated in human breast cancer tissues, and TINCR knockdown suppresses tumorigenesis in vitro and in vivo. Mechanistically, TINCR recruits DNMT1 to the miR-503-5p locus promoter, which increases the methylation and suppresses the transcriptional expression of miR-503-5p. Furthermore, TINCR also functions as a competing endogenous RNA to upregulate EGFR expression by sponging miR-503-5p. In addition, TINCR stimulates JAK2-STAT3 signaling downstream from EGFR, and STAT3 reciprocally enhances the transcriptional expression of TINCR. Our findings broaden the current understanding of the diverse manners in which TINCR functions in cancer biology. The newly identified STAT3-TINCR-EGFR-feedback loop could serve as a potential therapeutic target for human cancer.


Assuntos
Neoplasias da Mama/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Fator de Transcrição STAT3/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , DNA (Citosina-5-)-Metiltransferase 1/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Fator de Transcrição STAT3/genética
9.
Aging (Albany NY) ; 12(20): 20332-20349, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33080569

RESUMO

Breast cancer patients at the same stage may show different clinical prognoses or different therapeutic effects of systemic therapy. Differentially expressed genes of breast cancer were identified from GSE42568. Through survival, receiver operating characteristic (ROC) curve, random forest, GSVA and a Cox regression model analyses, genes were identified that could be associated with survival time in breast cancer. The molecular mechanism was identified by enrichment, GSEA, methylation and SNV analyses. Then, the expression of a key gene was verified by the TCGA dataset and RT-qPCR, Western blot, and immunohistochemistry. We identified 784 genes related to the 5-year overall survival time of breast cancer. Through ROC curve and random forest analysis, 10 prognostic genes were screened. These were integrated into a complex by GSVA, and high expression of the complex significantly promoted the recurrence-free survival of patients. In addition, key genes were related to immune and metabolic-related functions. Importantly, we identified methylation of MEX3A and TBC1D 9 and mutations events. Finally, the expression of UGCG was verified by the TCGA dataset and by experimental methods in our own samples. These results indicate that 10 genes may be potential biomarkers and therapeutic targets for long-term survival in breast cancer, especially UGCG.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Sobreviventes de Câncer , Perfilação da Expressão Gênica , Transcriptoma , Neoplasias da Mama/mortalidade , Neoplasias da Mama/terapia , Proteínas de Ligação ao Cálcio/genética , Metilação de DNA , Bases de Dados Genéticas , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Glucosiltransferases/genética , Humanos , Proteínas de Membrana/genética , Nomogramas , Fosfoproteínas/genética , Valor Preditivo dos Testes , Proteínas de Ligação a RNA/genética , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
10.
Signal Transduct Target Ther ; 5(1): 102, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32561709

RESUMO

Immunotherapy, designed to exploit the functions of the host immune system against tumors, has shown considerable potential against several malignancies. However, the utility of immunotherapy is heavily limited due to the low response rate and various side effects in the clinical setting. Immune escape of tumor cells may be a critical reason for such low response rates. Noncoding RNAs (ncRNAs) have been identified as key regulatory factors in tumors and the immune system. Consequently, ncRNAs show promise as targets to improve the efficacy of immunotherapy in tumors. However, the relationship between ncRNAs and tumor immune escape (TIE) has not yet been comprehensively summarized. In this review, we provide a detailed account of the current knowledge on ncRNAs associated with TIE and their potential roles in tumor growth and survival mechanisms. This review bridges the gap between ncRNAs and TIE and broadens our understanding of their relationship, providing new insights and strategies to improve immunotherapy response rates by specifically targeting the ncRNAs involved in TIE.


Assuntos
MicroRNAs/genética , Neoplasias/genética , RNA não Traduzido/genética , Evasão Tumoral/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imunoterapia/tendências , MicroRNAs/imunologia , Neoplasias/imunologia , RNA não Traduzido/imunologia , Evasão Tumoral/genética
11.
Cancer Immunol Res ; 8(7): 937-951, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32321773

RESUMO

Long noncoding RNAs (lncRNA) that are associated with immune checkpoints have not been identified, and the mechanism by which such lncRNAs might regulate the expression of immune checkpoints is unknown in human cancer. Immune checkpoint-associated lncRNAs (ICP-lncRNA) were identified and validated via a comprehensive bioinformatic analysis of The Cancer Genome Atlas data. These ICP-lncRNAs were involved in key immune response and immune cell receptor signaling pathways. The expression of ICP-lncRNAs was upregulated and correlated with a poor prognosis in patients with cancer. HLA complex P5 (HCP5) and myocardial infarction associated transcript (MIAT) promoted tumor growth and upregulated the expression of PD-L1/CD274 via a competing endogenous RNA mechanism of sponging miR-150-5p. The combination of MIAT knockdown and PD-L1 antibody administration showed a synergistic inhibitory effect on tumor growth. Finally, the expression of both HCP5 and MIAT was confirmed to be transcriptionally suppressed by CCCTC-binding factor (CTCF), and lipopolysaccharide induced CTCF eviction from the HCP5 and MIAT promoters, attenuating the transcriptionally suppressive activity of CTCF. This study enlarges the functional landscape of known lncRNAs in human cancer and indicates novel insights into their roles in the field of tumor immunity and immunotherapy. These findings may aid in the comprehensive management of human cancer with immunotherapy.


Assuntos
Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/imunologia , Neoplasias/genética , RNA Longo não Codificante/imunologia , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Humanos , Proteínas de Checkpoint Imunológico/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Prognóstico , RNA Longo não Codificante/genética , Regulação para Cima
12.
Cell Rep ; 29(4): 800-809.e5, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31644904

RESUMO

Cockayne syndrome (CS) is a rare genetic neurodevelopmental disorder, characterized by a deficiency in transcription-coupled subpathway of nucleotide excision DNA repair (TC-NER). Mutation of the Cockayne syndrome B (CSB) gene affects basal transcription, which is considered a major cause of CS neurologic dysfunction. Here, we generate a rat model by mimicking a nonsense mutation in the CSB gene. In contrast to that of the Csb-/- mouse models, the brains of the CSB-deficient rats are more profoundly affected. The cerebellar cortex shows significant atrophy and dysmyelination. Aberrant foliation of the cerebellum and deformed hippocampus are visible. The white matter displays high glial fibrillary acidic protein (GFAP) staining indicative of reactive astrogliosis. RNA sequencing (RNA-seq) analysis reveals that CSB deficiency affects the expression of hundreds of genes, many of which are neuronal genes, suggesting that transcription dysregulation could contribute to the neurologic disease seen in the CSB rat models.


Assuntos
Síndrome de Cockayne/genética , Códon sem Sentido , DNA Helicases/genética , Enzimas Reparadoras do DNA/genética , Modelos Animais de Doenças , Proteínas de Ligação a Poli-ADP-Ribose/genética , Animais , Células Cultivadas , Cerebelo/metabolismo , Cerebelo/patologia , Cerebelo/fisiopatologia , Síndrome de Cockayne/patologia , Síndrome de Cockayne/fisiopatologia , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Bainha de Mielina/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Ratos , Ratos Sprague-Dawley
13.
Am J Transl Res ; 10(3): 837-846, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29636873

RESUMO

OBJECTIVE: This study aims to observe the effect and mechanism of Xiaoru Sanjie Jiaonang (XRSJ) on the treatment of mammary gland hyperplasia, and provide a theoretical basis and clinical evidence for clinical expansion. METHODS: Japanese white rabbits were randomly divided into three groups: high-, middle- and low-dose groups; Xiaoyao Pill group; model control group; normal control group. The observation points were as follows: before XRSJ administration, three months after XRSJ administration, and three months after XRSJ discontinuance. Changes in breast height, morphological changes of the mammary gland under a light and electron microscope, and the expression of ki-67 were observed. At the same time, patients diagnosed with mammary gland hyperplasia at an Outpatient Clinic were selected and divided into treatment groups. These patients received XRSJ and Xiaoyao Pills, respectively, for one month, while patients in the control group did not receive any drug treatment. Clinical efficacy was observed while rechecking at the Outpatient Clinic after three months. Treatment with a therapeutic dose of XRSJ could significantly reduce breast height, decrease the number of lobules and acini in hyperplastic mammary glands and the layer number of ductal glandular epithelial cells, substantially lower the content of serum estradiol (E2), significantly downregulate the expression of ki-67 protein in mammary tissues, and inhibit mammary gland hyperplasia. CONCLUSION: XRSJ treatment can relieve mammary tissue hyperplastic lesions, reduce E2 levels and downregulate the expression of ki-67. It has a significant therapeutic effect on mammary gland hyperplasia.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...