Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Mol Carcinog ; 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39016636

RESUMO

LAIR1, a receptor found on immune cells, is capable of binding to collagen and is involved in immune-related diseases. However, the precise contribution of LAIR1 expressed on hepatocellular carcinoma (HCC) cells to tumor microenvironment is still unclear. In our study, bioinformatics analysis and immunofluorescence were employed to study the correlation between LAIR1 levels and clinical indicators. Transwell and scratch tests were used to evaluate how LAIR1 affected the migration and invasion of HCC cells. The chemotactic capacity and alternative activation of macrophages were investigated using RT-qPCR, transwell, and immunofluorescence. To investigate the molecular mechanisms, transcriptome sequencing analysis, Western blot, nucleus/cytoplasm fractionation, ELISA, and cytokine microarray were employed. We revealed a significant correlation between the presence of LAIR1 and an unfavorable outcome in HCC. We indicated that LAIR1 promoted migration and invasion of HCC cells through the AKT-IKKß-p65 axis. Additionally, the alternative activation and infiltration of tumor-associated macrophages induced by LAIR1 were reliant on the upregulation of IL6 and CCL5 within this axis, respectively. In conclusion, blocking LAIR1 was found to be an effective approach in combating the cancerous advancement of HCC.

2.
Cancer Immunol Immunother ; 72(12): 4103-4119, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37755466

RESUMO

Immunotherapy, which aims to enhance the function of T cells, has emerged as a novel therapeutic approach for hepatocellular carcinoma (HCC). Nevertheless, the clinical utility of using flow cytometry to assess immune cell infiltration (ICI) is hindered by its cumbersome procedures, prompting the need for more accessible methods. Here, we acquired gene expression profiles and survival data of HCC from TCGA and GSE10186 datasets. The patients were categorized into two clusters of ICI, and a set of 11 characteristic genes responsible for the differentiation performance of these ICI clusters were identified. Subsequently, we successfully developed a modified ICI score (mICIS) by utilizing the expression levels of these genes. The efficacy of our mICIS was confirmed via mass cytometry, flow cytometry, and immunohistochemistry. Our research indicated that the favorable overall survival (OS) rate could be attributed to the improved function of anti-tumor leukocytes rather than their infiltration. Furthermore, we observed that the low score group exhibited lower expression levels of T-cell exhaustion-associated genes, which was confirmed in both HCC tissues from patients and mice, which demonstrated that the benefits of the low scores were due to enhanced active/cytotoxic CD8+ T cells and reduced exhausted CD8+ T cells. Additionally, our mICIS stratified the benefits derived from immunotherapies. Lastly, we observed a misalignment between CD8+ T-cell infiltration and function in HCC. In summary, our mICIS demonstrated proficiency in assessing the OS rate of HCC and offering significant stratified data pertaining to distinct responses to immunotherapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animais , Camundongos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Linfócitos T CD8-Positivos , Imuno-Histoquímica , Imunoterapia , Microambiente Tumoral
3.
Cell Commun Signal ; 21(1): 80, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-37081450

RESUMO

BACKGROUND: Interleukin-33 (IL-33), defined as "alarming", exert diverse functions through signaling via the suppression of tumorigenicity 2 (ST2). However, the physiological roles of IL-33/ST2 signaling during acetaminophen (APAP)-induced liver injury are still poorly understood by modern medicine (AILI). This research aims to explore the relationship between IL-33/ST2 and stimulator of interferon (IFN) response cGAMP interactor 1 (STING)-mediated signal transduction. METHODS: C57BL/6N mice (WT) and IL-33-deficient mice (KO) were intraperitoneally injected with APAP (250 mg/kg). Recombinant IL-33 (500 ng/mouse) and the cGAS/STING inhibitor RU.521 (200 g/kg) were combined to treat AILI. For mechanistic research in vitro, CRISPR-mediated KD technology, immunoprecipitation, mass spectrometry, and immunofluorescence were utilized. RESULTS: We discovered that IL-33 deficient mice had increased APAP-induced hepatotoxicity, DNA accumulation, and type 1 IFN production. Mechanistic analysis revealed that IL-33/ST2 enhanced the interaction between Beclin-1 and STING, disrupting STING dimerization, IRF3 phosphorylation, nuclear transport, and IFN-1 gene transcription in HepaRG and Huh7 cells. Beclin-1 interacted with the C-terminus of STING, causing Lys338 acetylation and autophagy degradation of STING. ST2 depletion increased STING signal transduction and IFN-1 promoter activity. Surprisingly, the cGAS/STING inhibitor RU.521 and recombinant IL-33 together improved AILI in vivo. CONCLUSIONS: These results shed insight on the potential of inhibiting cGAS/STING as a therapy for AILI and emphasize the crucial role of IL-33/ST2 signaling in the regulation of APAP-induced STING signaling. Video Abstract.


Assuntos
Acetaminofen , Proteína 1 Semelhante a Receptor de Interleucina-1 , Animais , Camundongos , Acetaminofen/efeitos adversos , Autofagia , Proteína Beclina-1 , Imunidade Inata/genética , Interleucina-33 , Camundongos Endogâmicos C57BL , Nucleotidiltransferases/metabolismo , Transdução de Sinais
4.
Int J Exp Pathol ; 103(3): 102-111, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35429078

RESUMO

Emerging evidences have shown that long noncoding RNA (lncRNA) plays an important role in the immune escape of cancer cells. Our previous study has demonstrated that lncRNA MIAT is associated with the immune infiltration of hepatocellular carcinoma (HCC). However, the underlying mechanism of MIAT regulating the PD-L1-mediated immune escape of HCC is poorly understood. Quantitative real-time PCR (qRT-PCR) was used to detect the expression of MIAT and PD-L1 mRNA in HCC. The relationship between MIAT, miR-411-5p, STAT3 and PD-L1 was explored by dual-luciferase reporter assay, cytotoxicity assay, Western blot and RNA immunoprecipitation (RIP). In addition, the xenograft model was established to determine the effect of MIAT on PD-L1 expression in vivo. We found that MIAT and PD-L1 were significantly upregulated in HCC tissues and the expression of PD-L1 was regulated by MIAT. The knockdown of MIAT enhanced the cytotoxicity of T cells on HCC cells. MIAT negatively regulated miR-411-5p expression, upregulated STAT3 and ultimately increased PD-L1 expression from the transcription level. The inhibition of miR-411-5p reversed STAT3 and PD-L1 expression inhibited by MIAT knockdown in HCC cells. This study suggests a novel lncRNA-mediated mechanism for HCC cells to evade the immune response; MIAT/miR-411-5p/STAT3/PD-L1 may be a novel therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , RNA Longo não Codificante , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
5.
Cell Biol Toxicol ; 38(6): 1159-1173, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35438468

RESUMO

Recent studies have suggested that the initiation and progression of hepatocellular carcinoma (HCC) are closely associated with lipopolysaccharide (LPS) of intestinal bacteria. However, the role of LPS in immune regulation of HCC remains largely unknown. An orthotopic Hepa1-6 tumor model of HCC was constructed to analyze the effect of LPS on the expression of immune checkpoint molecules PD-1 and PD-L1. Then we verified the regulation of PD-L1 by LPS in HCC cells. Based on the previous finding that lncRNA MIR155HG regulates PD-L1 expression in HCC cells, we analyzed the relationship of LPS signaling pathway molecules with PD-L1 and MIR155HG by bioinformatics. The molecular mechanism of MIR155HG regulating PD-L1 expression induced by LPS was investigated by RNA pull-down followed by mass spectrometry, RNA immunoprecipitation, fluorescence in situ hybridization, and luciferase reporter assay. Finally, the HepG2 xenograft model was established to determine the role of MIR155HG on PD-L1 expression in vivo. We showed that LPS induced PD-1 and PD-L1 expression in mouse tumor tissues and induced PD-L1 expression in HCC cells. Mechanistically, upregulation of METTL14 by LPS promotes the m6A methylation of MIR155HG, which stabilizes MIR155HG relying on the "reader" protein ELAVL1 (also known as HuR)-dependent pathway. Moreover, MIR155HG functions as a competing endogenous RNA (ceRNA) to modulate the expression of PD-L1 by miR-223/STAT1 axis. Our results suggested that LPS plays a critical role in immune escape of HCC through METTL14/MIR155HG/PD-L1 axis. This study provides a new insight for understanding the complex immune microenvironment of HCC. 1. LPS plays a critical role in immune escape of HCC, especially HCC with cirrhosis. 2. Our study reveals that LPS regulates PD-L1 by m6A modification of lncRNA in HCC. 3. MIR155HG plays an important role in LPS induced PD-L1 expression. 4. LPS-MIR155HG-PD-L1 regulatory axis provides a new target for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , RNA Longo não Codificante , Humanos , Camundongos , Animais , Carcinoma Hepatocelular/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Lipopolissacarídeos/farmacologia , Neoplasias Hepáticas/patologia , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Hibridização in Situ Fluorescente , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Microambiente Tumoral
6.
Exp Cell Res ; 362(2): 504-514, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29253535

RESUMO

The HepG2 cell line is widely used in studying liver diseases because of its immortalization, but its clinical application is limited by its low expression of the urea synthesis key enzymes and cytochromes P450 (CYPs). On the basis of our previous work, we investigated the transcriptional regulation of arginase 1 (Arg1) and ornithine transcarbamylase (OTC) in HepG2 cells. We also screened for the optimal combination of liver enrichment transcription factors (LETFs) and xenobiotic nuclear receptors that can promote the expression of key urea synthases and five major CYPs in HepG2 cells. Thus, recombinant HepG2 cells were established. Results showed that C/EBPß, not C/EBPα, could upregulate expression of Arg1 and PGC1α and HNF4α cooperatively regulate the expression of OTC. The two optimal combinations C/EBPß+HNF4α+HNF6+PXR and C/EBPß+HNF4α+HNF6+CAR were selected. Compared with the control cells, the recombinant HepG2 cells modified by the two optimal combinations exhibited enhanced ammonia metabolism and CYP enzyme activity. Moreover, the HepG2/(C/EBPß+HNF4α+HNF6+PXR) cells more strongly reduced ammonia than any other combination tested in this study. The present work indicated that optimizing the combination of transcription factors will simultaneously promote hepatocyte ammonia metabolism and drug metabolism. The recombinant HepG2 liver cell line constructed by the optimal combination provided an improved alternative means for bioartificial liver applications and drug toxicity testing.


Assuntos
Amônia/farmacologia , Arginase/genética , Neoplasias Hepáticas/metabolismo , Ornitina Carbamoiltransferase/genética , Amônia/metabolismo , Arginase/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Inativação Metabólica/efeitos dos fármacos , Inativação Metabólica/genética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Regiões Promotoras Genéticas/genética
8.
J Enzyme Inhib Med Chem ; 34(1): 799-807, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30879343

RESUMO

mPGES-1 is a terminal rate-limiting enzyme responsible for inflammation-induced PGE2 production. The inhibition of mPGES-1 has been considered as a safe and effective target for the treatment of inflammation and cancer. However, a specific, efficient, and simple method for high-throughput screening of mPGES-1 inhibitors is still lacking. In this study, we developed a fluorescence imaging strategy to monitor the expression of mPGES-1 via CRISPR/Cas9 knock-in system. Immunofluorescence colocalisation, Sanger sequencing, RNAi, and IL-1ß treatment all confirmed the successful construction of mPGES-1 reporter cells. The fluorescence signal intensity of the reporter cells treated with four conventional mPGES-1 inhibitors was considerably attenuated via flow cytometry and fluorescent microplate reader, demonstrating that the reporter cells can be used as an efficient and convenient means for screening and optimising mPGES-1 inhibitors. Moreover, it provides a new technical support for the development of targeted small molecule compounds for anti-inflammatory and tumour therapy.


Assuntos
Sistemas CRISPR-Cas , Inibidores Enzimáticos/farmacologia , Fígado/efeitos dos fármacos , Prostaglandina-E Sintases/antagonistas & inibidores , Citometria de Fluxo , Fluorescência , Imunofluorescência , Células HEK293 , Células Hep G2 , Ensaios de Triagem em Larga Escala , Humanos , Interleucina-1beta/farmacologia , Fígado/citologia , Fígado/enzimologia , Prostaglandina-E Sintases/genética , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Bibliotecas de Moléculas Pequenas/farmacologia
10.
J Cell Mol Med ; 21(9): 2036-2045, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28272778

RESUMO

Carbamoyl phosphate synthase 1 (CPS1) is the rate-limiting enzyme in the first step of the urea cycle and an indispensable enzyme in the metabolism of human liver. However, CPS1 epigenetic regulation involves promoter analysis and the role of liver-enriched transcription factors (LETFs), which is not fully elucidated. In this work, the promoter region of hCPS1 gene was cloned, and its activity was investigated. An LETF, hepatocyte nuclear factor 3-beta (HNF3ß), was found to promote the transcriptional expression of CPS1 in liver-derived cell lines. In addition, dual-luciferase reporter assay shows that the essential binding sites of the HNF3ß may exist in the oligonucleotide -70 nt to +73 nt. Two putative binding sites are available for HNF3ß. Mutation analysis results show that the binding site 2 of HNF3ß was effective, and the transcriptional activity of CPS1 promoter significantly decreased after mutation. Electrophoretic mobile shift assay (EMSA) and ChIP assay confirmed that HNF3ß can interact with the binding site in the CPS1 promoter region of -70 nt to +73 nt promoter region in vivo and in vitro to regulate the transcription of CPS1. Moreover, HNF3ß overexpression enhanced the transcription of CPS1 and consequently improved the mRNA and protein levels of CPS1, whereas the knockdown of HNF3ß showed the opposite effects. Finally, urea production in cells was measured, and ammonia detoxification improved significantly in cells after transfection with HNF3ß. HNF3ß plays a vital role in regulation of CPS1 gene and could promote the metabolism of ammonia by regulating CPS1 expression.


Assuntos
Carbamoil-Fosfato Sintase (Amônia)/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fígado/citologia , Regiões Promotoras Genéticas , Amônia/metabolismo , Sequência de Bases , Sítios de Ligação , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Inativação Metabólica , Nucleotídeos/genética , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Transcrição Gênica , Ureia/metabolismo
11.
Cancer Sci ; 108(6): 1144-1156, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28295868

RESUMO

Several studies have produced contradictory findings about the prognostic implications for inhibitor of apoptosis proteins (IAP) in different types of cancer. Cellular inhibitor of apoptosis 2 (cIAP2/BIRC) is one of the most extensively characterized human IAP. To date, no studies have focused on the expression level of cIAP2 in human gallbladder cancer (GBC), and the mechanism of cIAP2 in GBC invasion and lymphangiogenesis remains unclear. Therefore, in the present study, cIAP2 expression in GBC was detected using quantitative real-time polymerase chain reaction and immunohistochemistry, and the relationship between cIAP2 levels in cancer tissues and the clinicopathological characteristics of patients was analyzed. The biological effect of cIAP2 in GBC cells was tested using the Cell Counting Kit-8 Assay, Transwell assays and the ability of human dermal lymphatic endothelial cells (HDLEC) to undergo tube formation. The role of cIAP2 in activating the NF-κB pathway was determined using a dual-luciferase reporter assay, immunofluorescence staining, western blotting and ELISA. Finally, an animal model was used to further confirm the role of cIAP2 in lymphangiogenesis. We showed that cIAP2 expression was elevated in human GBC tissues and correlated with a negative prognosis for patients. Moreover, cIAP2 was identified as a lymphangiogenic factor of GBC cells and, thus, promoted lymph node metastasis in GBC cells. Our study is the first to suggest that cIAP2 can promote GBC invasion and lymphangiogenesis by activating the NF-κB pathway.


Assuntos
Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/patologia , Proteínas Inibidoras de Apoptose/genética , Linfangiogênese/genética , NF-kappa B/genética , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases/genética , Idoso , Animais , Proteína 3 com Repetições IAP de Baculovírus , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Metástase Linfática/genética , Metástase Linfática/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Prognóstico
12.
Mediators Inflamm ; 2015: 372750, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26538827

RESUMO

We investigated the mechanism of caffeine in influencing HBx(+) hepatocytes to synthesize PGE2. The inhibitory effect of caffeine on hepatocyte proliferation increased with increasing caffeine concentrations (200-800 µM) and treatment times (1-7 days), which was first observed at the second test time point (caffeine treatment for 4 days). The inhibition of caffeine on the growth of HL7702-HBx and HepG2-HBx cells was most obvious at 800 µM caffeine and at caffeine treatment for 7 days. The PGE2 secretion and the expression of mPGES-1 and EGR1 were downregulated, whereas PPARγ expression was upregulated. The mPGES-1 promoter activity of HBx(+) hepatocytes decreased more significantly than that of HBx(-) hepatocytes. Moreover, the expression of EGR1 and PPARγ changed more significantly in HBx(+) hepatocytes cultured for 12 to 24 hours in the presence of 5 mM caffeine. This limited success may be attributed to caffeine releasing the binding of HBx and PPARγ and furthermore affecting the mPGES-1 expression by EGR1 in HBx(+) hepatocytes. The results indicate that caffeine could effectively reduce PGE2 synthesis in HBx(+) hepatocytes by specifically blocking the PPARγ-EGR1-mPGES-1 pathway, thereby providing a new evidence of molecular biology for the hypothesis that drinking coffee is beneficial to HBV-infected patients.


Assuntos
Cafeína/química , Dinoprostona/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Hepatócitos/metabolismo , Hepatócitos/virologia , Oxirredutases Intramoleculares/metabolismo , Fígado/metabolismo , Linhagem Celular , Proliferação de Células , Regulação para Baixo , Regulação da Expressão Gênica , Células Hep G2 , Hepatócitos/citologia , Humanos , Fígado/efeitos dos fármacos , Inibidores de Fosfodiesterase/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Prostaglandina-E Sintases , Transativadores/metabolismo , Proteínas Virais Reguladoras e Acessórias
13.
Cell Physiol Biochem ; 34(4): 1152-65, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25277242

RESUMO

BACKGROUND AND AIM: Receptor interacting protein(RIP)-1 is thought to have a significant role in inflammation signaling pathways; however, the role of RIP-1 in malignant tumors is largely unknown. METHODS: The present study examined the functions and underlying mechanisms of RIP-1 in gallbladder cancer in vitro and in vivo. In this study we determined the expression and role of RIP-1 in 60 clinical specimens from patients with gallbladder cancer and 3 gallbladder cancer cell lines. Using siRNA targeting RIP-1, plasmid vectors (phU6-EGFP-puro/siRIP-1) were constructed and transfected into the gallbladder cells to characterize the biological effect of RIP-1. Results : In vitro experiments indicated that silencing of RIP-1 in NOZ cells significantly suppressed growth and invasion. Furthermore, silencing of RIP-1 affected the RIP1-NF-κB/c-jun(AP-1)-VEGF-C pathways in NOZ cells. Silencing of RIP-1 in vivo inhibited tumor growth in a NOZ cell subcutaneous xenograft model. Immunohistochemstry analysis of the tumor in thesubcutaneous xenograft model also suggested that RIP-1 mediates the expression of VEGF-C. CONCLUSION: We have elucidated therelationship between RIP-1 overexpression and the growth and invasion of gallbladder cancer from clinical specimens using a xenograft model. We provide evidence that a reduction in the expression of RIP-1 in gallbladder cancer cells can exert inhibitory effects on the ability of cells to grow and invade in vitro. Thus, targeting RIP-1may be useful in the treatment of gallbladder cancer.


Assuntos
Carcinoma/genética , Proliferação de Células/genética , Neoplasias da Vesícula Biliar/genética , Invasividade Neoplásica/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas de Ligação a RNA/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Feminino , Neoplasias da Vesícula Biliar/patologia , Humanos , Masculino , Pessoa de Meia-Idade , NF-kappa B/genética , Invasividade Neoplásica/patologia , Transdução de Sinais/genética , Fator de Transcrição AP-1/genética , Fator C de Crescimento do Endotélio Vascular
14.
Exp Dermatol ; 23(7): 512-3, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24816403

RESUMO

In this study, we observed that rhFNHN29 and rhFNHC36, two recombinant heparin-binding domain polypeptides of fibronectin, suppressed adhesion and invasion of B16F10 and A375 melanoma cells mediated by integrin αv and α2 in a dose-dependent manner. Combined with low-concentration epirubicin (EPI), rhFNHN29 or rhFNHC36 exhibited a synergistic inhibition on the viability and metastasis of B16F10 cells. Moreover, in the presence of high-concentration rhFNHN29 or rhFNHC36, the Ets-1 activity and the expression of p-FAK, p-Erk1/2 and Ets-1 were notably downregulated in B16F10 cells. Ets-1 is one of the central regulatory links for rhFNHN29 and rhFNHC36 to suppress the adhesion and invasion of melanoma cells. Combining rhFNHN29 or rhFNHC36 with EPI may be a good way to alleviate invasiveness or chemoresistance in melanoma.


Assuntos
Fibronectinas/metabolismo , Heparina/metabolismo , Melanoma/patologia , Peptídeos/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Animais , Adesão Celular , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Humanos , Masculino , Melanoma Experimental , Camundongos , Invasividade Neoplásica , Ligação Proteica , Estrutura Terciária de Proteína
15.
Microbiol Spectr ; 12(1): e0286023, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38032223

RESUMO

IMPORTANCE: Hepatitis B virus (HBV)-specific CD8+ T cells play a central role in the clearance of virus and HBV-related liver injury. Acute infection with HBV induces a vigorous, multifunctional CD8+ T cell response, whereas chronic one exhibits a weaker response. Our study elucidated HBV-specific T cell responses in terms of viral abundance rather than the timing of infection. We showed that in the premalignant stage, the degree of impaired T cell function was not synchronized with the viral surface antigen, which was attributed the liver's tolerance to the virus. However, after the development of hepatocellular carcinoma, T cell exhaustion was inevitable, and it was marked by the exhaustion of the signature transcription factor TOX.


Assuntos
Hepatite B Crônica , Hepatite B , Humanos , Vírus da Hepatite B , Hepatite B Crônica/patologia , Linfócitos T CD8-Positivos , Antígenos Virais
16.
Cell Signal ; 115: 111039, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38199599

RESUMO

BACKGROUND: An increasing number of studies have reported the involvement of oncogenes in the regulation of the immune system. LAIR1 is an immunosuppressive molecule and its role in immune-related diseases has been mainly reported. To date, it is unclear whether LAIR1 in tumor cells is involved in immune regulation. Therefore, the aim of this study was to investigate the role of LAIR1 in the immune microenvironment of hepatocellular carcinoma (HCC) to seek the novel therapeutic discoveries. METHODS: Tumor Immune Dysfunction and Exclusion database was used to predict the response of LAIR1 expression to immune checkpoint blockade. CD8+ T cells were co-cultured with HCC cells, and the killing efficiency of leukocytes on HCC cells was detected by flow cytometry. Flow cytometry was also used to detect the expression of inhibitory receptors. In addition, Western blot, immunofluorescence, and nucleus/cytoplasm fractionation experiments were performed to explore the molecular mechanisms by which LAIR1 created a suppressive tumor microenvironment. RESULTS: LAIR1 expression in HCC was associated with worse immune prognosis and T-cell dysfunction. HCC cells overexpressing LAIR1 co-cultured with CD8+ T cells induced exhaustion of latter. Mechanism studies indicated that LAIR1 in HCC cells up-regulated the phosphorylation of ß-catenin by inducing the phosphorylation of GSK-3ß, leading to the impairment of the expression and the nuclear localization signal of ß-catenin. Low ß-catenin expression and nuclear localization signal inhibited MYC-mediated PD-L1 expression. Therefore, PD-L1 up-regulated by LAIR1 caused the exhaustion of infiltrating CD8+ T cells in HCC, which aggravated the malignant progression of HCC. CONCLUSION: LAIR1 increased PD-L1 expression through the GSK-3ß/ß-catenin/MYC/PD-L1 pathway and promoted immune evasion of HCC cells. Targeted inhibition of LAIR1 helped to enhance the immune killing effect of CD8+ T cells in HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Antígeno B7-H1/metabolismo , beta Catenina/metabolismo , Sinais de Localização Nuclear/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
17.
Int Immunopharmacol ; 136: 112415, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38850791

RESUMO

The microenvironment of hepatocellular carcinoma (HCC) is characterized by hypoxia, which leads to immune evasion of HCC. Therefore, gaining a comprehensive understanding of the mechanism underlying the impact of hypoxia on HCC cells may provide valuable insights into immune checkpoint therapy. Based on analysis of databases and clinical samples, we observed that expression level of programmed cell death ligand 1 (PD-L1) and long non-coding RNA (lncRNA) MIR155HG in patients in the hypoxia group were higher than those in the non-hypoxia group. Furthermore, there was a positive correlation between the expression of PD-L1 and MIR155HG with that of HIF-1α. In vitro experiments using hypoxic treatment demonstrated an increase in PD-L1 and MIR155HG expression levels in HCC cells. While the hypoxia-induced upregulation of PD-L1 could be reversed by knocking down MIR155HG. Mechanistically, as a transcription factor, HIF-1α binds to the promoter region of MIR155HG to enhance its transcriptional activity under hypoxic conditions. Hypoxia acts as a stressor promoting nuclear output of ILF3 leading to increased binding of ILF3 to MIR155HG, thereby enhancing stability for HIF-1α mRNA. In vivo, knocking down MIR155HG inhibit subcutaneous tumor growth, reduce the expression of HIF-1α and PD-L1 within tumors; additionally, it enhances anti-tumor immunity response. These findings suggested that through inducing MIR155HG to interact with ILF3, hypoxia increases HIF-1α mRNA stability resulting in elevated PD-L1 expression in HCC and thus promoting immune escape. In summary, this study provides new insights into the effects of hypoxia on HCC immunosuppression.


Assuntos
Antígeno B7-H1 , Carcinoma Hepatocelular , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias Hepáticas , Estabilidade de RNA , RNA Longo não Codificante , Animais , Feminino , Humanos , Masculino , Camundongos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Evasão Tumoral/genética , Microambiente Tumoral/imunologia
18.
J Cell Biochem ; 114(5): 1097-104, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23161342

RESUMO

This study investigated the molecular mechanisms of liver cells with HBx expression on epithelium-mesenchymal transition (EMT) change using Western blot analysis and Transwell assay to assess EMT-related protein expression and cell mobility. Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were used to test the Twist promoter containing different STAT3 binding loci. Electrophoretic mobility band-shift assay (EMSA) was used to detect Twist activity. Results showed that HBx expression affected the EMT-related protein expression and the cell mobility of liver cancer cells (MHCC97) and liver cells (HL-7702) in vitro or in vivo. These proteins exhibited reversed expression to a certain extent after Twist inhibition. In addition, the wound-healing capability and the mobility of HL-7702/HBx cells were lower than those treated with control-siRNA. The expressions of p-STAT3 and Twist were positively correlated with HBx expression. The second STAT-3 binding sequence in the Twist promoter region of the HL-7702/HBx cells was the first locus. Twist activity in the HL-7702/HBx2 cells was higher than that in HL-7702 cells. Moreover, the activity decreased when the cells were treated with HBx-siRNA to inhibit HBx expression, or with STAT3 inhibitor to reduce STAT3 activation. Therefore, Twist is essential for the regulation of the mobility of liver cells with HBx expression. HBx activates the Twist promoter by activating STAT3 and promotes EMT occurrence in liver cells.


Assuntos
Transição Epitelial-Mesenquimal , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/patologia , Proteínas Nucleares/metabolismo , Fator de Transcrição STAT3/metabolismo , Transativadores/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Linhagem Celular , Movimento Celular , Hepatócitos/virologia , Humanos , Fígado/metabolismo , Fígado/virologia , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , Proteínas Nucleares/antagonistas & inibidores , RNA Interferente Pequeno/metabolismo , Proteína 1 Relacionada a Twist/antagonistas & inibidores , Proteínas Virais Reguladoras e Acessórias
19.
Hepatobiliary Pancreat Dis Int ; 12(5): 525-32, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24103284

RESUMO

BACKGROUND: Key enzyme deficiency in the dual-pathway of ammonia metabolism leads to low detoxification capacity of HepG2 cells. Previously, we established a HepG2/AFhGS cell line with overexpression of human glutamine synthetase (hGS) in pathway 1 and a HepG2/(hArgI+hOTC)4 cell line with overexpression of human arginase I (hArgI) and human ornithine transcarbamylase (hOTC) in pathway 2. The present study aimed to investigate whether simultaneous recovery of the two pathways contributes to the further improvement of ammonia detoxification in HepG2 cells. METHODS: We adopted a recombinant retrovirus carrying the hGS gene to infect HepG2/(hArgI+hOTC)4 cells and selected a new recombinant HepG2 cell line. The capacities of ammonia tolerance and detoxification in cells were detected by biochemical methods. Cell cycle PCR chip was used to assess the changes of gene expression. RESULTS: Introducing hGS into HepG2/(hArgI+hOTC)4 cells did not lead to hGS overexpression, but inhibited hArgI expression. The levels of synthetic glutamine and urea in HepG2/(hArgI+hOTC+AFhGS)1 cells were significantly lower than those in HepG2/(hArgI+hOTC)4 cells when cultured in the medium with 10 and 15 mmol/L glutamate (Glu) and with 60 and 180 mmol/L NH4Cl, respectively. In addition, the comparison of different cell growth showed that HepG2/AFhGS cells significantly lagged behind the other cells by the 5th and 7th day, indicating that introduction of hGS impedes HepG2 cell proliferation. Analysis of the mechanism suggested that the decreased expression of BCL2 played an important role. CONCLUSIONS: This study demonstrated that the recovery of two ammonia metabolic pathways in HepG2 cells is not helpful in increasing ammonia metabolism. The reinforcement of the pathway of urea metabolism is more important and valuable in improving the ammonia metabolism capacity in HepG2 cells.


Assuntos
Amônia/metabolismo , Neoplasias Hepáticas/metabolismo , Arginase/genética , Arginase/metabolismo , Biotransformação , Ciclo Celular , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glutamato-Amônia Ligase/genética , Glutamato-Amônia Ligase/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Ornitina Carbamoiltransferase/genética , Ornitina Carbamoiltransferase/metabolismo , Transfecção , Ureia/metabolismo
20.
J Cell Commun Signal ; 17(4): 1131-1143, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37878185

RESUMO

Interleukin-33 (IL-33) (NF-HEV), a chromatin-associated nuclear cytokine, is a member of the IL-1 family. IL-33 possesses a nuclear localization signal and a homeodomain (a structure resembling a helix-turn-helix) that can bind to nuclear chromatin. Research has revealed that IL-33 can function as a nuclear factor to regulate various biological processes. This review discusses the cellular localization, functional effects, and immune regulation of full length IL-33 (FLIL-33), cytokine IL-33 (sIL-33) and nuclear IL-33 (nIL-33). In addition, the post-translational modifications of nIL-33 and the hypothesis of using nIL-33 as a treatment method were also summarized. A multidisciplinary approach is required which integrates methods and techniques from genomics, proteomics, cell biology and immunology to provide comprehensive insights into the function and therapeutic potential of nIL-33.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA