Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Radiat Oncol Biol Phys ; 119(1): 219-233, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-37914138

RESUMO

PURPOSE: Radiation therapy is a vital adjuvant treatment for liver cancer, although the challenge of radiation-induced liver diseases (RILDs) limits its implementation. Kupffer cells (KCs) are a crucial cell population of the hepatic immune system, and their biologic function can be modulated by multiple epigenetic RNA modifications, including N6-methyladenosine (m6A) methylation. However, the mechanism for m6A methylation in KC-induced inflammatory responses in RILD remains unclear. The present study investigated the function of m6A modification in KCs contributing to RILD. METHODS AND MATERIALS: Methylated RNA-immunoprecipitation sequencing and RNA transcriptome sequencing were used to explore the m6A methylation profile of primary KCs isolated from mice after irradiation with 3 × 8 Gy. Western blotting and quantitative real-time PCR were used to evaluate gene expression. DNA pulldown and chromatin immunoprecipitation assays were performed to verify target gene binding and identify binding sites. RESULTS: Methylated RNA-immunoprecipitation sequencing revealed significantly increased m6A modification levels in human KCs after irradiation, suggesting the potential role of upregulated m6A in RILD. In addition, the study results corroborated that methyltransferase-like 3 (METTL3) acts as a main modulator to promote the methylation and gene expression of TEAD1, leading to STING-NLRP3 signaling activation. Importantly, it was shown that IGF2BP2 functions as an m6A "reader" to recognize methylated TEAD1 mRNA and promote its stability. METTL3/TEAD1 knockdown abolished the activation of STING-NLRP3 signaling, protected against RILD, and suppressed inflammatory cytokines and hepatocyte apoptosis. Moreover, clinical human normal liver tissue samples collected after irradiation showed increased expression of STING and interleukin-1ß in KCs compared with nonirradiated samples. Notably, STING pharmacologic inhibition alleviated irradiation-induced liver injury in mice, indicating its potential therapeutic role in RILD. CONCLUSIONS: The results of our study reveal that TEAD1-STING-NLRP3 signaling activation contributes to RILD via METTL3-dependent m6A modification.


Assuntos
Células de Kupffer , Neoplasias Hepáticas , Humanos , Camundongos , Animais , Células de Kupffer/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Regulação para Cima , Piroptose , Neoplasias Hepáticas/metabolismo , RNA Mensageiro/genética , Metiltransferases/genética , Proteínas de Ligação a RNA/fisiologia
2.
Cancer Lett ; 582: 216507, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38048841

RESUMO

Although Poly (ADP-ribose) polymerase (PARP) inhibitors have been clinically approved for cancers with BRCA mutations and are known to augment radiotherapy responses, their roles in promoting the abscopal effect and mediating immunotherapy in BRCA-proficient hepatocellular carcinoma (HCC) remain underexplored. Our study elucidates that olaparib enhances the radio-sensitivity of HCC cells. Coadministration of olaparib and irradiation induces significant DNA damage by generating double-strand breaks (DSBs), as revealed both in vitro and in immune-deficient mice. These DSBs activate the cGAS-STING pathway, initiating immunogenic cell death in abscopal tumors. STING activation reprograms the immune microenvironment in the abscopal tumors, triggering the release of type I interferon and chemokines, including CXCL9, CXCL10, CXCL11, and CCL5. This in turn amplifies T cell priming against tumor neoantigens, leading to an influx of activated, neoantigen-specific CD8+ T-cells within the abscopal tumors. Furthermore, olaparib attenuated the immune exhaustion induced by radiation and enhances the responsiveness of HCC to immune checkpoint inhibitors. Collectively, our data advocate that a synergistic regimen of PARP inhibitors and radiotherapy can strategically reinforce both local (primary) and systemic (abscopal) tumor control, bolstering HCC susceptibility to immunotherapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Piperazinas , Animais , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/radioterapia , Linfócitos T CD8-Positivos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/radioterapia , Neoplasias Hepáticas/metabolismo , Ftalazinas/farmacologia , Linhagem Celular Tumoral , Microambiente Tumoral
3.
Radiat Res ; 200(6): 531-537, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38014555

RESUMO

Patients with hepatobiliary tumors who accept radiotherapy are at risk for radiation-induced liver fibrosis. MicroRNAs (miRNAs) have been implicated in the pathogenesis of radiation-induced liver damage and possess potential as novel biomarkers and therapeutic targets. However, the role of miR-146a-5p in radiation-induced liver fibrosis is less well understood. The current study was designed to evaluate the role of miR-146a-5p in radiation-induced liver fibrosis in mice and to investigate the possible mechanisms involved in miR-146a-5p-mediated effects. The experiments were performed on Institute of Cancer Research (ICR) mice which received fractionated radiation (30 Gy in 5 fractions) to the liver. The results show radiation could induce histopathological changes, liver dysfunction and fibrosis accompanied with decreased miR-146a-5p expression. miR-146a-5p agomir treatment resulted in recovery of liver function and reduced the amount of alpha-smooth muscle actin (α-SMA), collagen 1, protein tyrosine phosphatase receptor type A (PTPRA) and phosphorylated SRC in the livers of irradiated mice. Therefore, our study reveals that miR-146a-5p inhibits the progression of hepatic fibrosis after radiation treatment. And the beneficial role of miR-146a-5p may be relevant to PTPRA-SRC signaling pathway.


Assuntos
MicroRNAs , Humanos , Camundongos , Animais , MicroRNAs/genética , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Fibrose , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores
4.
Int J Radiat Oncol Biol Phys ; 112(5): 1243-1255, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34986380

RESUMO

PURPOSE: Radiation therapy (RT) is one of the main treatments for patients with unresectable hepatocellular carcinoma (HCC). Emerging evidence indicates that the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) stimulator of interferon gene (STING) pathway is crucial in RT-induced antitumor immune responses. Here, we discovered that activation of the cancer cell-intrinsic cGAS-STING pathway mediated immune cloaking after RT-induced DNA damage. METHODS AND MATERIALS: Key regulatory proteins in the cGAS-STING signaling pathway in human and murine HCC cell lines were knocked out or down using CRISPR and CRISPR-associated protein 9 or small interfering RNA. The underlying mechanism of immune cloaking and clinical significance of cGAS-STING-induced programmed cell death ligand 1 (PD-L1) expression were studied with both ex vivo analyses and in vitro experiments. RESULTS: RT upregulated PD-L1 in patients with HCC, which correlated with poor survival. RT activated cGAS-STING, increasing immune-checkpoint PD-L1 expression in human and mouse liver cancer cells. Ionizing radiation activated the STING-TANK-binding kinase 1 (TBK1)-interferon regulatory factor 3 (IRF3) innate immune pathway, leading to PD-L1 upregulation in HCC cells and inhibiting cytotoxic T-lymphocyte activity and protecting tumor cells from immune-mediated eradication. Knockdown of cGAS, STING, TBK1, and IRF3 reversed the antitumor effect of cytotoxic T-lymphocyte-mediated cytotoxicity after ionizing radiation in vitro or in vivo. RT potentiated the antitumor effect of programmed cell death protein 1 and PD-L1 axis blockade and augmented cytotoxic T-cell (CTL) infiltration in HCC tumors in immunocompetent mice. CD8 depletion compromised the synergetic antitumor effect of combined RT and anti-PD-L1 blockade, demonstrating that CD8+ CTLs are required for antitumor immunity induced by combination therapy. CONCLUSIONS: Our results identified an immune-cloaking mechanism for RT-activated, innate immune cGAS-STING and suggested that RT enhances HCC immunotherapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas de Membrana , Nucleotidiltransferases , Animais , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/radioterapia , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/radioterapia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Regulação para Cima
5.
Mol Cancer Ther ; 21(1): 16-24, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34670783

RESUMO

TGFß is a pleiotropic cytokine that plays critical roles to define cancer cell phenotypes, construct the tumor microenvironment, and suppress antitumor immune responses. As such, TGFß is a lynchpin for integrating cancer cell intrinsic pathways and communication among host cells in the tumor and beyond that together affect responses to genotoxic, targeted, and immune therapy. Despite decades of preclinical and clinical studies, evidence of clinical benefit from targeting TGFß in cancer remains elusive. Here, we review the mechanisms by which TGFß acts to oppose successful cancer therapy, the reported prognostic and predictive value of TGFß biomarkers, and the potential impact of inhibiting TGFß in precision oncology. Paradoxically, the diverse mechanisms by which TGFß impedes therapeutic response are a principal barrier to implementing TGFß inhibitors because it is unclear which TGFß mechanism is functional in which patient. Companion diagnostic tools and specific biomarkers of TGFß targeted biology will be the key to exploiting TGFß biology for patient benefit.


Assuntos
Neoplasias/tratamento farmacológico , Fator de Crescimento Transformador beta/uso terapêutico , Humanos , Prognóstico , Transdução de Sinais , Fator de Crescimento Transformador beta/farmacologia
6.
Front Mol Biosci ; 8: 655361, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262937

RESUMO

Acute kidney injury (AKI) is a severe and frequently observed condition associated with high morbidity and mortality. The molecular mechanisms underlying AKI have not been elucidated due to the complexity of the pathophysiological processes. Thus, we investigated the key biological molecules contributing to AKI based on the transcriptome profile. We analyzed the RNA sequencing data from 39 native human renal biopsy samples and 9 reference nephrectomies from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) and Gene Ontology (GO) analysis revealed that various GO terms were dysregulated in AKI. Gene set enrichment analysis (GSEA) highlighted dysregulated pathways, including "DNA replication," "chemokine signaling pathway," and "metabolic pathways." Furthermore, the protein-to-protein interaction (PPI) networks of the DEGs were constructed, and the hub genes were identified using Cytoscape. Moreover, weighted gene co-expression network analysis (WGCNA) was performed to validate the DEGs in AKI-related modules. Subsequently, the upregulated hub genes STUB1, SOCS1, and VHL were validated as upregulated in human AKI and a mouse cisplatin-induced AKI model. Moreover, the biological functions of STUB1 were investigated in renal tubular epithelial cells. Cisplatin treatment increased STUB1 expression in a dose-dependent manner at both the mRNA and protein levels. Knockdown of STUB1 by siRNA increased the expression of proapoptotic Bax and cleaved caspase-3 while decreasing antiapoptotic Bcl-2. In addition, silencing STUB1 increased the apoptosis of HK-2 cells and the proinflammatory cytokine production of IL6, TNFα, and IL1ß induced by cisplatin. These results indicated that STUB1 may contribute to the initiation and progression of AKI by inducing renal tubular epithelial cell apoptosis and renal inflammation.

7.
Int J Radiat Oncol Biol Phys ; 111(2): 491-501, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34044094

RESUMO

PURPOSE: Radiation therapy, which is vital for the treatment of primary liver cancer, comes with unavoidable liver injury, which limits its implementation. N6-methyladenosine (m6A) methylation is involved in many molecular functions. However, its role in radiation-induced liver diseases (RILD) remains unknown. Herein, we investigate the role of m6A methylation in RILD. METHODS AND MATERIALS: Methylated RNA-immunoprecipitation sequencing and RNA transcriptome sequencing were used to reveal the methylation pattern of human hepatic stellate cells (HSCs) exposed to irradiation. C3H/HeN mice and stimulator of interferon genes (STING)-deficient mice underwent x-ray irradiation of 24 Gy in 3 fractions. The m6A methylation of the high-mobility group box 1 (HMGB1) transcript was validated using methylated RNA immunoprecipitation, RNA immunoprecipitation, luciferase assays, and a messenger RNA decay assay. RESULTS: Human hepatic stellate cells showed significant differences in methylation patterns after 8 Gy of x-ray irradiation. Irradiation recruited AlkB homolog 5 (ALKBH5) to demethylate m6A residues in the 3' untranslated region of HMGB1, which resulted in the activation of STING-interferon regulatory factor 3 signaling. Changes in the transcription of the 3' untranslated region of HMGB1 occurred after the knockdown of ALKBH5, which were eliminated after m6A residue mutation. Strikingly, ALKBH5 deficiency or HMGB1 silencing both attenuated type I interferon production and decreased hepatocyte apoptosis. In vivo depletion of ALKBH5 abolished the upregulation of HMGB1-mediated STING signaling and decreased liver inflammation, which was consistent with STING-/- mice treated with irradiation. Notably, YTHDF2 (m6A reader protein) directly bound to HMGB1 m6A-modified sites and promoted its degradation. CONCLUSIONS: ALKBH5-dependent HMGB1 expression mediates STING-interferon regulatory factor 3 innate immune response in RILD.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase/fisiologia , Proteína HMGB1/fisiologia , Células Estreladas do Fígado/efeitos da radiação , Imunidade Inata/efeitos da radiação , Hepatopatias/etiologia , Neoplasias Hepáticas/radioterapia , Proteínas de Membrana/fisiologia , Animais , Proteína HMGB1/genética , Humanos , Interferon Tipo I/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteínas de Ligação a RNA/fisiologia , Transdução de Sinais/fisiologia
8.
Chemosphere ; 271: 129831, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33736218

RESUMO

High-resolution transmission electron microscopy observations of bastnäsite from the Maoniuping rare earth element (REE) deposit at Panxi, SW China, revealed the presence of nanoparticles in the surface of bastnäsite crystal. The nanoparticles are identified as the bastnäsite nanocrystals, with 5-30 nm in length, by energy-dispersive spectrometry and fast fourier transform patterns. This represents the first observation of bastnäsite nanoparticle in nature, confirming a new form of migration and precipitation of La and Ce in the hydrothermal fluids. Meanwhile, we herein reveal that random attachment of small bastnäsite nanoparticles during aggregation-based growth initially produces large bastnäsite nanoparticles, driven by the tectonic stress. Subsequent evolution of the random aggregation and orientation of large bastnäsite nanoparticles result in bastnäsite crystal formation, driven by the tectonic stress and inherent surface stress. This underlines the bastnäsite crystallization by random aggregation and the significant role of tectonic stress in forming the bastnäsite crystal in REE deposit.


Assuntos
Metais Terras Raras , Nanopartículas , China , Cristalização , Metais Terras Raras/análise , Microscopia Eletrônica de Transmissão
9.
Cell Mol Immunol ; 18(7): 1718-1728, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32203191

RESUMO

Liver damage upon exposure to ionizing radiation (IR), whether accidental or therapeutic, can contribute to liver dysfunction. Currently, radiotherapy (RT) is used for various cancers including hepatocellular carcinoma (HCC); however, the treatment dose is limited by radiation-induced liver disease (RILD) with a high mortality rate. Furthermore, the precise molecular mechanisms of RILD remain poorly understood. Here, we investigated RILD pathogenesis using various knockout mouse strains subjected to whole-liver irradiation. We found that hepatocytes released a large quantity of double-stranded DNA (dsDNA) after irradiation. The cGAS-STING pathway in non-parenchymal cells (NPCs) was promptly activated by this dsDNA, causing interferon (IFN)-I production and release and concomitant hepatocyte damage. Genetic and pharmacological ablation of the IFN-I signaling pathway protected against RILD. Moreover, clinically irradiated human peri-HCC liver tissues exhibited substantially higher STING and IFNß expression than non-irradiated tissues. Increased serum IFNß concentrations post-radiation were associated with RILD development in patients. These results delineate cGAS-STING induced type 1 interferon release in NPCs as a key mediator of IR-induced liver damage and described a mechanism of innate-immunity-driven pathology, linking cGAS-STING activation with amplification of initial radiation-induced liver injury.


Assuntos
Carcinoma Hepatocelular , Doença Hepática Crônica Induzida por Substâncias e Drogas , Interferon Tipo I , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/radioterapia , DNA/metabolismo , Humanos , Interferon Tipo I/metabolismo , Neoplasias Hepáticas/radioterapia , Proteínas de Membrana/metabolismo , Camundongos , Transdução de Sinais
10.
Med Sci Monit ; 26: e922378, 2020 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-32449506

RESUMO

BACKGROUND The aim of this study was to construct a nomogram to predict the prognosis of patients with gastrointestinal stromal tumor (GIST). MATERIAL AND METHODS We enrolled 4086 GIST patients listed in the SEER database from 1998 to 2015. They were separated to 2 groups: an experimental group (n=2862) and a verification group (n=1224). A nomogram was constructed by using statistically significant prognostic factors. RESULTS A nomogram that included age, sex, marital status, tumor location, grade, SEER stage, tumor size, and surgical management was developed. It can be used to predict overall survival (OS), while adding AJCC 7th TNM stage can predict cancer-specific survival (CSS). The C-index used to forecast OS and CSS nomograms was 0.778 (95% CI, 0.76-0.79) and 0.818 (95% CI, 0.80-0.84), respectively. CONCLUSIONS The nomogram can effectively predict 3- and 5-year CSS in patients with GIST, and its use can improve clinical practice.


Assuntos
Tumores do Estroma Gastrointestinal/mortalidade , Idoso , Feminino , Tumores do Estroma Gastrointestinal/patologia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Gradação de Tumores , Estadiamento de Neoplasias , Nomogramas , Prognóstico , Modelos de Riscos Proporcionais , Programa de SEER
11.
J Cell Biochem ; 121(11): 4431-4439, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32003059

RESUMO

Lung cancer is one of the fatal tumors. The tumor microenvironment plays a key role in regulating tumor progression. To figure out the role of tumor microenvironment in lung adenocarcinoma (LUAD), ESTIMATE algorithm was used to evaluate the immune scores in LUAD. Patients with low immune scores had a worse overall survival (OS) compared with high immune scores. Using RNA-Seq data of 489 patients in The Cancer Genome Atlas (TCGA), differentially expressed genes (DEGs) were identified between high- and low-immune score groups. Based on the DEGs, nine-gene signature was constructed by the least absolute shrinkage and selection operator Cox regression model in TCGA set. The signature demonstrated significant prognostic value in both TCGA and Gene Expression Omnibus database. Multivariate Cox regression analyses indicated that nine-genes signature was an independent prognostic factor. Subgroup analysis also revealed a robust prognostic ability of nine-gene signature. A nomogram with a C-index of 0.722 had a favorable power for predicting 3-, 5-, and 10-year survival for clinical use by integrating nine-gene signature and other clinical features. Co-expression and functional enrichment analysis showed that nine-gene signature was significantly associated with immune response and provided potential profound molecules for revealing the mechanism of tumor initiation and progression. In conclusion, we revealed the significance of immune infiltration and built a novel nine-gene signature as a reliable prognostic factor for patients with LUAD.


Assuntos
Adenocarcinoma de Pulmão/mortalidade , Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/mortalidade , Nomogramas , Células Estromais/imunologia , Microambiente Tumoral/imunologia , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/imunologia , Adenocarcinoma de Pulmão/patologia , Idoso , Biomarcadores Tumorais/imunologia , Feminino , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Masculino , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Transcriptoma
12.
J Cell Physiol ; 235(11): 8270-8282, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31960423

RESUMO

The role of circular RNA (circRNA) in radiation-induced liver disease (RILD) remains largely unknown. In this study, Ras-related C3 botulinum toxin substrate 1 (RAC1) was elevated in irradiated human hepatic stellate cell (HSC) line LX2, the important effector cell mediating RILD. Overexpression of RAC1 promotes cell proliferation, proinflammatory cytokines production, and α-smooth muscle actin expression, which were blocked by microRNA (miR)-146a-5p mimics. CircRNA RSF1 (circRSF1) was upregulated in irradiated LX2 cells and predicted to harbor binding site for miR-146a-5p. Biotinylated-RNA pull down and dual-luciferase reporter detection confirmed the direct interaction of circRSF1 and miR-146a-5p. Enforced expression of circRSF1 increased RAC1 expression by acting as miR-146a-5p sponge to inhibit miR-146a-5p activity, and thus enhanced the cell viability, and promoted inflammatory and fibrotic phenotype of irradiated LX2 cells. These findings indicate a functional regulatory axis composing of circRSF1, miR-146a-5p, and RAC1 in irradiated HSC, which may provide attractive therapeutic targets for RILD.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Células Estreladas do Fígado/efeitos da radiação , Cirrose Hepática/genética , MicroRNAs/genética , Proteínas Nucleares/genética , Lesões por Radiação/genética , Transativadores/genética , Linhagem Celular , Células Estreladas do Fígado/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Cirrose Hepática/etiologia , Fenótipo , RNA Circular/genética , Proteínas rac1 de Ligação ao GTP/biossíntese
13.
Front Oncol ; 9: 896, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552194

RESUMO

Background and Objective: Radiation-induced lymphopenia has a tangible impact on overall survival (OS) in multiple solid tumors. We investigated the association between circulating lymphocyte populations (CLPs) before and after stereotactic body radiation therapy (SBRT) and OS in patients with hepatocellular carcinoma (HCC). Materials and Methods: Seventy-eight HCC patients treated with SBRT between January 2013 and June 2017 were retrospectively analyzed. Baseline and post-treatment total peripheral lymphocyte counts (TPLCs) and values of different CLPs were obtained and analyzed for clinical outcomes. Univariate and multivariate Cox regression analyses were used to explore the independent prognostic factors for patient survival. Results: The one-, two- and three-year OS rates were 94.8, 75.9, and 63.3%, respectively. The mean TPLCs before and 10 days after SBRT were 1.4 × 109/L and 0.7 × 109/L, respectively. The TPLC recovered to its baseline value 1 year after SBRT. Multivariate analysis results revealed that variables, including tumor necrosis factor-alpha (TNF-α) level <5.5 ng/mL and post-treatment TPLC <0.45 × 109/L were independent factors for inferior OS. Further analysis showed that the values of CLPs, including CD3+, CD4+, CD8+, CD19+, and CD16+56+ cells dropped profoundly 10 days after SBRT, among which CD19+ B cell count was mostly depleted and gradually recovered after 2 months. Univariate analysis showed that both baseline and post-treatment TPLC and CLP (except post-treatment B cell) counts were significantly associated with patient OS (p < 0.05 for each). Further stratified analysis performed according to OS at 2 years demonstrated that the CD16+CD56+ NK cell counts remained significantly elevated in patients with better survival (OS > 2 years) compared to those in short-term survivors at 10 days, 1 month, and 2 months after SBRT (p < 0.05 for each). In addition, there were significant differences in TPLC and CD8+ T cell counts in patients with long-term and short-term OS at 2 months after SBRT (p < 0.05). Conclusions: Peripheral lymphopenia after SBRT might be an independent prognostic factor for poorer outcome in HCC patients. Post-treatment lymphocyte subsets, including CD8+ T cell and NK cell counts were also associated with 2-year OS rates.

14.
Radiat Res ; 192(6): 621-629, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31560641

RESUMO

MicroRNAs (miRNAs) have been shown to play a pivotal role in the pathogenesis and maintenance of liver fibrosis by altering expression of their downstream target genes. However, their role in radiation-induced liver fibrosis has not been assessed in detail. Here, we investigated the role of miR-146a-5p and the target gene in regulation of fibrosis-related markers in the human hepatic stellate cell line LX2. LX2 cells were stimulated with 8 Gy of X rays and various concentrations of TGF-ß1 (0-5 ng/ml). Expression of α-SMA, collagen 1 and miR-146a-5p was evaluated. The MiR-146a-5p target gene predictions were performed using bioinformatics analysis and confirmed by dual-luciferase reporter experiment. The effect of miR-146a-5p and the involved target gene on the expression of these fibrogenic molecules was also assessed. Expression of α-SMA and collagen 1 were upregulated in response to radiation and/or TGF-ß1 treatment and miR-146a-5p levels were altered in LX2 cells. Restoration of miR-146a-5p expression suppressed expression of α-SMA and collagen 1 in irradiated and TGF-ß1-treated LX2 cells. Subsequent mechanism experiments revealed that miR-146a-5p overexpression inhibited PTPRA expression by binding to its 3'-untrans-lated region and reduced SRC activation. In addition, enhancement of PTPRA partially reversed the suppressive effect of miR-146a-5p on α-SMA and collagen 1 expression in LX2 cells. In conclusion, miR-146a-5p may negatively regulate the PTPRA-SRC signaling to inhibit expression of fibrosis-related markers in irradiated and TGF-ß1-stimulated LX2 cells.


Assuntos
Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , MicroRNAs/genética , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia , Regiões 3' não Traduzidas , Actinas/metabolismo , Linhagem Celular , Proliferação de Células , Colágeno/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células Estreladas do Fígado/efeitos da radiação , Humanos , Cirrose Hepática/radioterapia , Raios X , Quinases da Família src/metabolismo
15.
Mol Cancer ; 18(1): 120, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31349837

RESUMO

BACKGROUND: Bone metastasis (BM) has long been recognized as a major threat to the quality of life of hepatocellular cancer (HCC) patients. While LncRNA34a (Lnc34a) has been shown to regulate colon cancer stem cell asymmetric division, its effect on HCC BM remains unknown. METHODS: In situ hybridization and quantitative real-time polymerase chain reaction (qRT-PCR) were used to detect the expression of Lnc34a in HCC tissues and cell lines. Ventricle injection model was constructed to explore the effect of Lnc34a on BM in vivo. The methylation of miR-34a promoter and histones deacetylation were examined by using bisulfate-sequencing PCR and chromatin immunoprecipitation assays. RNA pull down and RNA immunoprecipitation were performed to investigated the interaction between Lnc34a and epigenetic regulators. Dual-luciferase reporter assay was conducted to find miR-34a target. The involvement of TGF-ß pathway in the BM from HCC was determined by qRT-PCR, western, and elisa assays. RESULTS: We found that Lnc34a was significantly overexpressed in HCC tissues and associated with BM. Both in vitro and in vivo experiments indicate that the restoration or knockdown of Lnc34a expression in HCC cells had a marked effect on cellular migration, invasion, and metastasis. Mechanistic analyses suggested that Lnc34a epigenetically suppresses miR-34a expression through recruiting DNMT3a via PHB2 to methylate miR-34a promoter and HDAC1 to promote histones deacetylation. On the other hand, miR-34a targets Smad4 via the TGF-ß pathway, followed by altering the transcription of the downstream genes (i.e., CTGF and IL-11) that are associated with BM. CONCLUSIONS: Our study is the first to document the pro-bone metastatic role of Lnc34a in BM of HCC and reveal a novel mechanism for the activation of the TGF-ß signaling pathway in HCC BM, providing evidence of a potential therapeutic strategy in HCC BM.


Assuntos
Neoplasias Ósseas/secundário , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , RNA Longo não Codificante/genética , Adulto , Idoso , Animais , Biomarcadores , Neoplasias Ósseas/diagnóstico , Carcinoma Hepatocelular/diagnóstico por imagem , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Proibitinas , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
16.
Cancer Manag Res ; 11: 10929-10937, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32099457

RESUMO

PURPOSE: The neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are inflammatory indexes that may reflect immune response to tumors and prognosis. We investigated the prognostic values of pre-treatment and post-treatment NLR and PLR and changes in those ratios in patients with small hepatocellular carcinoma (sHCC) treated with stereotactic body radiation therapy (SBRT). PATIENTS AND METHODS: Sixty patients who received SBRT were retrospectively reviewed. NLR and PLR were calculated by division of neutrophil and platelet counts, respectively, by lymphocyte counts. Independent factors for progression-free survival (PFS) and overall survival (OS) were determined by the Kaplan-Meier method, log-rank test, and Cox multivariate regression. Hazard ratios (HRs) and 95% confidence intervals (CIs) were also calculated. RESULTS: The median follow-up was 36.9 (range: 4.1-73.5) months. Median PFS was 21.4 (range: 1.8-66.9) months. The 1-year and 2-year PFS rates were 76.7% and 55.0%, respectively. The 1-year and 2-year OS rates were 95.0% and 78.3%, respectively. In multivariate analysis, post-treatment PLR ≥263.0 indicated both poor PFS (HR: 3.70; 95% CI: 1.07-12.76, p=0.038) and OS (HR: 3.23; 95% CI: 1.01-9.11, p=0.043) for sHCC patients treated with SBRT. In addition, the presence of hepatitis infection and a low level of red blood cell count were also proved to be significantly associated with patients' poor prognosis (p<0.05 for each). Post-treatment increase in NLR ≥2.7-fold was shown to be a negative independent predictor of inferior OS (HR: 3.43; 95% CI: 1.14-10.38, p=0.029). CONCLUSION: High post-treatment PLR and change in NLR ≥2.7-fold were associated with poor prognosis in patients treated with SBRT and might be considered as reliable and independent prognostic biomarkers for patients with sHCC.

17.
Mol Med Rep ; 18(2): 2079-2087, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29956796

RESUMO

As a significant cause of mortality and morbidity, alcoholic liver disease (ALD) has been widely investigated. However, little is known about the underlying metabolic mechanisms involved in the complicated pathological processes of ALD. The present study used label­free quantitative proteomics and bioinformatics analyses to investigate the differentially expressed proteins (DEPs) and their functions in the livers of alcohol­feed (AF) and control pair­feed (PF) mice. As a result, 87 upregulated DEPs and 133 downregulated DEPs were identified in AF liver tissues compared with PF livers. Gene ontology and Kyoto encyclopedia of genes and genomes bioinformatics analyses demonstrated that the DEPs were significantly enriched in 'protein binding', 'metabolism', 'signal conduction' and 'immune response'. The expression of several core proteins including thyroid hormone receptor interactor 12 (TRIP12), NADH dehydrogenase (ubiquinone)1 α subcomplex, assembly factor 3 (NDUFAF3) and guanine monophosphate synthetase (GMPS) was validated by reverse transcription­quantitative polymerase chain reaction (RT­qPCR) in a larger series of samples. The RT­qPCR results confirmed that TRIP12, NDUFAF3 and GMPS genes were significantly differentially expressed in between the AF and PF samples. These results extend our understanding of the molecular mechanisms underlying the occurrence and development of ALD. The present study indicated that the majority of DEPs serve vital roles in multiple metabolic pathways and this extends our knowledge of the molecular mechanisms involved in the occurrence and progression of ALD.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Biologia Computacional , Hepatopatias Alcoólicas/metabolismo , Proteínas Mitocondriais/metabolismo , Proteômica , Ubiquitina-Proteína Ligases/metabolismo , Animais , Consumo Excessivo de Bebidas Alcoólicas/patologia , Modelos Animais de Doenças , Hepatopatias Alcoólicas/patologia , Masculino , Camundongos
18.
Cell Death Dis ; 9(2): 175, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29415990

RESUMO

Noncoding RNAs plays an important role in hepatocellular carcinoma (HCC). Here, we show that miR-124 was downregulated in HCC tissues and that the ectopic expression of miR-124 inhibited the proliferation and migration of HCC cells. We proposed that aquaporin 3 (AQP3) is a direct target of miR-124. AQP3 was upregulated in HCC tissues and inversely correlated with miR-124 expression. The overexpression of miR-124 decreased AQP3 expression. Indeed, AQP3 overexpression promoted cell proliferation and migration, whereas miR-124 knockdown suppressed cell proliferation and migration. Furthermore, we found that circular RNA HIPK3 (circHIPK3) acted as a miR-124 sponge and regulated the expression of the miR-124 target gene AQP3. circHIPK3 was upregulated in HCC tissues and positively correlated with AQP3 expression. Thus, silencing circHIPK3 inhibited cell proliferation and migration by downregulating AQP3 expression. Moreover, miR-124 inhibition rescued circHIPK3 knockdown induced reduction in cell proliferation and migration, as well as AQP3 expression. In vivo experiments also confirmed that circHIPK3 regulated xenograft tumor growth via the miR-124-AQP3 axis. These observations indicate a possible novel therapeutic strategy involving circular RNAs in HCC.


Assuntos
Aquaporina 3/genética , Carcinoma Hepatocelular/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , RNA/metabolismo , Animais , Aquaporina 3/metabolismo , Sequência de Bases , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , RNA/genética , RNA Circular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Regulação para Cima/genética
19.
Onco Targets Ther ; 10: 5783-5794, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29255366

RESUMO

Circular RNAs (circRNAs) are a novel class of noncoding RNAs that play an important role in cancer. However, the mechanisms by which circRNAs regulate gene expression in pancreatic ductal adenocarcinoma (PDAC) remain unclear. This study seeks to elucidate the role that circRNAs play in the proliferation of PDAC cells. On the basis of previous studies of circRNA expression profiles in PDAC, we found that the circRNA_100782 was markedly upregulated in PDAC tissue. Functional experiments revealed that circRNA_100782 down-regulation inhibited BxPC3 cell proliferation and colony formation. Loss-of-function studies showed that knockdown of circRNA_100782 inhibited cell proliferation by downregulating the microRNA-124 (miR-124) target genes interleukin-6 receptor (IL6R) and signal transducer and activator of transcription 3 (STAT3). Overexpression of miR-124 also inhibited BxPC3 cell proliferation by reducing the expression of IL6R and STAT3, which was consistent with the result of silencing circRNA_100782. In addition, luciferase assay revealed that miR-124 was a direct target of circRNA_100782. Silencing STAT3 inhibited BxPC3 cell proliferation and colony formation. Cell viability was reduced in BxPC3 cells treated with si-circRNA_100782 and miR-124 mimic, and this effect could be attenuated by activating STAT3. In vivo study validated that circRNA_100782 knockdown suppressed BxPC3 xenografts in nude mice. Taken together, these results suggest that circRNA_100782 regulates BxPC3 cell proliferation by acting as miR-124 sponge through the IL6-STAT3 pathway.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...