Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cancer Commun (Lond) ; 44(3): 384-407, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38407942

RESUMO

BACKGROUND: Liver cancer is a malignancy with high morbidity and mortality rates. Serpin family E member 2 (SERPINE2) has been reported to play a key role in the metastasis of many tumors. In this study, we aimed to investigate the potential mechanism of SERPINE2 in liver cancer metastasis. METHODS: The Cancer Genome Atlas database (TCGA), including DNA methylation and transcriptome sequencing data, was utilized to identify the crucial oncogene associated with DNA methylation and cancer progression in liver cancer. Data from the TCGA and RNA sequencing for 94 pairs of liver cancer tissues were used to explore the correlation between SERPINE2 expression and clinical parameters of patients. DNA methylation sequencing was used to detect the DNA methylation levels in liver cancer tissues and cells. RNA sequencing, cytokine assays, immunoprecipitation (IP) and mass spectrometry (MS) assays, protein stability assays, and ubiquitination assays were performed to explore the regulatory mechanism of SERPINE2 in liver cancer metastasis. Patient-derived xenografts and tumor organoid models were established to determine the role of SERPINE2 in the treatment of liver cancer using sorafenib. RESULTS: Based on the public database screening, SERPINE2 was identified as a tumor promoter regulated by DNA methylation. SERPINE2 expression was significantly higher in liver cancer tissues and was associated with the dismal prognosis in patients with liver cancer. SERPINE2 promoted liver cancer metastasis by enhancing cell pseudopodia formation, cell adhesion, cancer-associated fibroblast activation, extracellular matrix remodeling, and angiogenesis. IP/MS assays confirmed that SERPINE2 activated epidermal growth factor receptor (EGFR) and its downstream signaling pathways by interacting with EGFR. Mechanistically, SERPINE2 inhibited EGFR ubiquitination and maintained its protein stability by competing with the E3 ubiquitin ligase, c-Cbl. Additionally, EGFR was activated in liver cancer cells after sorafenib treatment, and SERPINE2 knockdown-induced EGFR downregulation significantly enhanced the therapeutic efficacy of sorafenib against liver cancer. Furthermore, we found that SERPINE2 knockdown also had a sensitizing effect on lenvatinib treatment. CONCLUSIONS: SERPINE2 promoted liver cancer metastasis by preventing EGFR degradation via c-Cbl-mediated ubiquitination, suggesting that inhibition of the SERPINE2-EGFR axis may be a potential target for liver cancer treatment.


Assuntos
Neoplasias Hepáticas , Serpina E2 , Humanos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Serpina E2/genética , Serpina E2/metabolismo , Sorafenibe , Ubiquitinação
2.
Adv Sci (Weinh) ; 9(29): e2201931, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36026578

RESUMO

Pancreatic ductal adenocarcinoma (PDAC), one of the worst prognosis types of tumors, is characterized by dense extracellular matrix, which compresses tumor vessels and forms a physical barrier to inhibit therapeutic drug penetration and efficacy. Herein, losartan, an antihypertension agent, is applied as a tumor stroma modulator and developed into a nanosystem. A series of lipophilic losartan prodrugs are constructed by esterification of the hydroxyl group on losartan to fatty acids. Based on the self-assembly ability and hydrodynamic diameter, the losartan-linoleic acid conjugate is selected for further investigation. To improve the stability in vivo, nanoassemblies are refined with PEGylation to form losartan nanoblocker (Los NB), and administered via intravenous injection for experiments. On murine models of pancreatic cancer, Los NB shows a greater ability to remodel the tumor microenvironment than free losartan, including stromal depletion, vessel perfusion increase, and hypoxia relief. Furthermore, Los NB pretreatment remarkably enhances the accumulation and penetration of 7-ethyl-10-hydroxycamptothecin (SN38)-loaded nanodrugs (SN38 NPs) in tumor tissues. Expectedly, overall therapeutic efficacy of SN38 NPs is significantly enhanced after Los NB pretreatment. Since losartan is one of the most commonly used antihypertension agents, this study may provide a potential for clinical transformation in stroma-rich PDAC treatment.


Assuntos
Antineoplásicos , Carcinoma Ductal Pancreático , Nanopartículas , Neoplasias Pancreáticas , Pró-Fármacos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Linhagem Celular Tumoral , Ácidos Graxos/uso terapêutico , Irinotecano/uso terapêutico , Ácidos Linoleicos/uso terapêutico , Losartan/farmacologia , Losartan/uso terapêutico , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Perfusão , Pró-Fármacos/uso terapêutico , Microambiente Tumoral , Neoplasias Pancreáticas
3.
Cancer Res ; 82(21): 3987-4000, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36043912

RESUMO

Liver cancer is characterized by aggressive growth and high mortality. Asialoglycoprotein receptor 1 (ASGR1), which is expressed almost exclusively in liver cells, is reduced in liver cancer. However, the specific mechanism of ASGR1 function in liver cancer has not been fully elucidated. On the basis of database screening, we identified ASGR1 as a tumor suppressor regulated by DNA methylation. Expression of ASGR1 was downregulated in liver cancer and correlated with tumor size, grade, and survival. Functional gain and loss experiments showed that ASGR1 suppresses the progression of liver cancer in vivo and in vitro. RNA sequencing and mass spectrometry showed that ASGR1 inhibits tyrosine phosphorylation of STAT3 by interacting with Nemo-like kinase (NLK). NLK bound the SH2 domain of STAT3 in an ATP-dependent manner and competed with glycoprotein 130 (GP130), ultimately suppressing GP130/JAK1-mediated phosphorylation of STAT3. ASGR1 altered the binding strength of NLK and STAT3 by interacting with GP130. Furthermore, the domain region of NLK was crucial for binding STAT3 and curbing its phosphorylation. Collectively, these results confirm that ASGR1 suppresses the progression of liver cancer by promoting the binding of NLK to STAT3 and inhibiting STAT3 phosphorylation, suggesting that approaches to activate the ASGR1-NLK axis may be a potential therapeutic strategy in this disease. SIGNIFICANCE: ASGR1 downregulation by DNA methylation facilitates liver tumorigenesis by increasing STAT3 phosphorylation.


Assuntos
Neoplasias Hepáticas , Humanos , Receptor de Asialoglicoproteína/genética , Receptor de Asialoglicoproteína/metabolismo , Receptor gp130 de Citocina , Neoplasias Hepáticas/patologia , Fator de Transcrição STAT3/metabolismo , Fosforilação , Domínios de Homologia de src , Proteínas Serina-Treonina Quinases
4.
Bioact Mater ; 18: 164-177, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35387168

RESUMO

A personalized medication regimen provides precise treatment for an individual and can be guided by pre-clinical drug screening. The economical and high-efficiency simulation of the liver tumor microenvironment (TME) in a drug-screening model has high value yet challenging to accomplish. Herein, we propose a simulation of the liver TME with suspended alginate-gelatin hydrogel capsules encapsulating patient-derived liver tumor multicellular clusters, and the culture of patient-derived tumor organoids(PDTOs) for personalized pre-clinical drug screening. The hydrogel capsule offers a 3D matrix environment with mechanical and biological properties similar to those of the liver in vivo. As a result, 18 of the 28 patient-derived multicellular clusters were successfully cultured as PDTOs. These PDTOs, along with hepatocyte growth factor (HGF) of non-cellular components, preserve stromal cells, including cancer-associated fibroblasts (CAFs) and vascular endothelial cells (VECs). They also maintain stable expression of molecular markers and tumor heterogeneity similar to those of the original liver tumors. Drugs, including cabazitaxel, oxaliplatin, and sorafenib, were tested in PDTOs. The sensitivity of PDTOs to these drugs differs between individuals. The sensitivity of one PDTO to oxaliplatin was validated using magnetic resonance imaging (MRI) and biochemical tests after oxaliplatin clinical treatment of the corresponding patient. Therefore, this approach is promising for economical, accurate, and high-throughput drug screening for personalized treatment.

5.
Oncogene ; 41(22): 3118-3130, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35477750

RESUMO

Targeting cytokinesis can suppress tumor growth by blocking cell division and promoting apoptosis. We aimed to characterize key cytokinesis regulator in hepatocellular carcinoma (HCC) progression, providing insights into identifying promising HCC therapeutic targets. The unbiased bioinformatic screening identified Anillin actin binding protein (ANLN) as a critical cytokinesis regulator involved in HCC development. Functional assay demonstrated that knockdown of ANLN inhibited HCC growth by inducing cytokinesis failure and DNA damage, leading to multinucleation and mitotic catastrophe. Mechanistically, ANLN acts as a scaffold to strengthen interaction between RACGAP1 and PLK1. ANLN promotes PLK1-mediated RACGAP1 phosphorylation and RhoA activation to ensure cytokinesis fidelity. To explore the function of ANLN in HCC tumorigenesis, we hydrodynamically transfected c-Myc and NRAS plasmids into Anln+/+, Anln+/-, and Anln-/- mice through tail vein injection. Hepatic Anln ablation significantly impaired c-Myc/NRAS-driven hepatocarcinogenesis. Moreover, enhanced hepatic polyploidization was observed in Anln ablation mice, manifesting as increasing proportion of cellular and nuclear polyploidy. Clinically, ANLN is upregulated in human HCC tissues and high level of ANLN is correlated with poor patients' prognosis. Additionally, the proportion of cellular polyploidy decreases during HCC progression and ANLN level is significantly correlated with cellular polyploidy proportion in human HCC samples. In conclusion, ANLN is identified as a key cytokinesis regulator contributing to HCC initiation and progression. Our findings revealed a novel mechanism of ANLN in the regulation of cytokinesis to promote HCC tumorigenesis and growth, suggesting targeting ANLN to inhibit cytokinesis may be a promising therapeutic strategy for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Proteínas Contráteis , Citocinese/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Proteínas dos Microfilamentos/metabolismo , Poliploidia
6.
Biochem Pharmacol ; 188: 114494, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33684390

RESUMO

Targeting the cell cycle checkpoints and DNA damage response are promising therapeutic strategies for cancer. Adavosertib is a potent inhibitor of WEE1 kinase, which plays a critical role in regulating cell cycle checkpoints. However, the effect of adavosertib on hepatocellular carcinoma (HCC) treatment, including sorafenib-resistant HCC, has not been thoroughly studied. In this study, we comprehensively investigated the efficacy and pharmacology of adavosertib in HCC therapy. Adavosertib effectively inhibited the proliferation of HCC cells in vitro and suppressed tumor growth in HCC xenografts and patient-derived xenograft (PDX) models in vivo. Additionally, adavosertib treatment effectively inhibited the motility of HCC cells by impairing pseudopodia formation. Further, we revealed that adavosertib induced DNA damage and premature mitosis entrance by disturbing the cell cycle. Thus, HCC cells accumulating DNA damage underwent mitosis without G2/M checkpoint arrest, thereby leading to mitotic catastrophe and apoptosis under adavosertib administration. Given that sorafenib resistance is common in HCC in clinical practice, we also explored the efficacy of adavosertib in sorafenib-resistant HCC. Notably, adavosertib still showed a desirable inhibitory effect on the growth of sorafenib-resistant HCC cells. Adavosertib markedly induced G2/M checkpoint arrest and cell apoptosis in a dose-dependent manner, confirming the similar efficacy of adavosertib in sorafenib-resistant HCC. Collectively, our results highlight the treatment efficacy of adavosertib in HCC regardless of sorafenib resistance, providing insights into exploring novel strategies for HCC therapy.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Proteínas de Ciclo Celular/antagonistas & inibidores , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Hepáticas/tratamento farmacológico , Fenótipo , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirazóis/administração & dosagem , Pirimidinonas/administração & dosagem , Animais , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Inibidores Enzimáticos/administração & dosagem , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteínas Tirosina Quinases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
7.
Theranostics ; 11(5): 2318-2333, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33500727

RESUMO

Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide, and its specific mechanism has not been fully elucidated. Inactivation of tumor suppressors may contribute to the occurrence, progression, and recurrence of HCC. DNA methylation is a crucial mechanism involved in regulating the occurrence of HCC. Herein, we aimed to identify the key methylation-related tumor suppressors as well as potential biomarkers and therapeutic targets in HCC. Methods: Combined analysis of TCGA and GEO databases was performed to obtain potential methylation-related tumor suppressors in HCC. Methyl-target sequencing was performed to analyze the methylation level of the GNA14 promoter. The diagnostic value of GNA14 as a predictor of HCC was evaluated in HCC tumor samples and compared with normal tissues. The functional role of GNA14 and its upstream and downstream regulatory factors were investigated by gain-of-function and loss-of-function assays in vitro. Subcutaneous tumorigenesis, lung colonization, and orthotopic liver tumor model were performed to analyze the role of GNA14 in vivo.Results: The expression of GNA14 was found to be downregulated in HCC and it was negatively correlated with hepatitis B virus (HBV) infection, vascular invasion, and prognosis of HCC. DNA methylation was demonstrated to be responsible for the altered expression of GNA14 and was regulated by HBV-encoded X protein (HBx). GNA14 regulated the RB pathway by promoting Notch1 cleavage to inhibit tumor proliferation, and might inhibit tumor metastasis by inhibiting the expression of JMJD6. Conclusion: GNA14 could be regulated by HBx by modulating the methylation status of its promoter. We identified GNA14 as a potential biomarker and therapeutic target for HCC.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/patologia , Metilação de DNA , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica , Hepatite B/complicações , Neoplasias Hepáticas/patologia , Animais , Apoptose , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virologia , Proliferação de Células , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hepatite B/virologia , Vírus da Hepatite B/isolamento & purificação , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Camundongos , Camundongos Nus , Prognóstico , Regiões Promotoras Genéticas , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Hazard Mater ; 403: 123598, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-32768862

RESUMO

A new method to solidify Cr(VI), Cr(III), and Ni(II) simultaneously by using a glass-ceramics microstructure was used. For a crystallization temperature of 870 °C, base glass (air cooling) with a mass ratio of CaO / SiO2 (R(C/S)) of 6/16, 10/16 precipitated a CaNiSi2O6 crystalline phase that could not solidify Ni(II) stably, and with an increase in R(C/S), the CaNiSi2O6 crystal precipitation tendency increased. When R(C/S) = 6/16, the crystallization temperature was 760 °C and spinel that was enriched in chromium and nickel formed preferentially during the inhibition of CaNiSi2O6 formation, indicating that lowering R(C/S) can inhibit the formation of CaNiSi2O6. Cr and Ni were mostly solidified in spinel, some of them enter augite (Ca (Mg, Fe, Al) (Si, Al)2O6) phases, a few of them were distributed in the glass matrix. The results of toxic leaching showed that the leached concentration of total Cr and Ni2+ in spinel glass-ceramics were 0 ppm and 0.07 ppm respectively, and the spinel glass-ceramics for simultaneous solidification of Cr(VI), Cr(III) and Ni(II) had excellent curing effect.

9.
Oncogene ; 40(1): 163-176, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33097858

RESUMO

Ether-à-go-go-1 (EAG1), one of the potassium channels, is involved in various physiological processes and plays an important role in the tumorigenesis of many kinds of cancer. EAG1 is highly expressed in hepatocarcinoma cells and is closely related to clinical prognosis, but the molecular mechanism remains elusive. In this study, we verified that EAG1 promotes the proliferation of hepatocellular carcinoma (HCC) both in vitro and in vivo. It promotes cell cycle progression by inhibiting the ubiquitination of SKP2. In addition, EAG1 promotes the migration and invasion of HCC by promoting cell pseudopod formation. Furthermore, in a high-pressure plasmid-injected mouse liver orthotopic carcinoma model, astemizole, an EAG family blocker, can significantly inhibit the formation of liver cancer. Meanwhile, liver-specific EAG1 knockout mice show resistance to hepatocarcinogenesis. This research demonstrated that EAG1 plays an important role in the progression of HCC, and could be a potential therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Neoplasias Hepáticas/patologia , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Prognóstico , Ubiquitinação , Regulação para Cima
10.
Neurology ; 95(24): e3355-e3363, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33028663

RESUMO

OBJECTIVE: To determine the effect of remote ischemic postconditioning (RIPC) on patients with acute ischemic stroke (AIS) undergoing IV thrombolysis (IVT). METHODS: A single-center randomized controlled trial was performed with patients with AIS receiving IVT. Patients in the RIPC group were administered RIPC treatment (after IVT) during hospitalization. The primary endpoint was a score of 0 or 1 on the modified Rankin scale (mRS) at day 90. The safety, tolerability, and neuroprotection biomarkers associated with RIPC were also evaluated. RESULTS: We collected data from both the RIPC group (n = 34) and the control group (n = 34). The average duration of hospitalization was 11.2 days. There was no significant difference between 2 groups at admission for the NIH Stroke Scale score (p = 0.364) or occur-to-treatment time (p = 0.889). Favorable recovery (mRS score 0-1) at 3 months was obtained in 71.9% of patients in the RIPC group vs 50.0% in the control group (adjusted odds ratio 9.85, 95% confidence interval 1.54-63.16; p = 0.016). We further found significantly lower plasma S100-ß (p = 0.007) and higher vascular endothelial growth factor (p = 0.003) levels in the RIPC group than in the control group. CONCLUSIONS: Repeated RIPC combined with IVT can significantly facilitate recovery of nerve function and improve clinical prognosis of patients with AIS. CLINICALTRIALSGOV IDENTIFIER: NCT03218293. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that RIPC after tissue plasminogen activator treatment of AIS significantly increases the proportion of patients with an MRS score of 0 or 1 at 90 days.


Assuntos
Fibrinolíticos/administração & dosagem , Pós-Condicionamento Isquêmico , AVC Isquêmico/terapia , Avaliação de Resultados em Cuidados de Saúde , Administração Intravenosa , Idoso , Terapia Combinada , Feminino , Humanos , Pós-Condicionamento Isquêmico/métodos , AVC Isquêmico/sangue , AVC Isquêmico/tratamento farmacológico , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Subunidade beta da Proteína Ligante de Cálcio S100/sangue , Ativador de Plasminogênio Tecidual/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/sangue
11.
Int J Oncol ; 57(1): 161-170, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32377719

RESUMO

Optimizing the currently available treatment options for pancreatic cancer (PC) is a priority. Cabazitaxel (CTX), a semisynthetic taxane, is mainly used for treating patients with PC who are resistant to paclitaxel (PTX) or docetaxel, due its poor affinity for P­glycoprotein. However, there are only a few studies demonstrating the effect of CTX on PC. The present study aimed to investigate the efficiency and underlying mechanism of CTX in PC treatment. Cell proliferation, colony formation assay and apoptosis analysis were achieved in the two human PC cell lines AsPC­1 and BxPC­3. Drug sensitivity test was performed in BxPC­3 tumor­bearing mice. The results demonstrated that CTX had a lower half maximal inhibitory concentration compared with PTX for the inhibition of cell proliferation, both in vivo and in vitro. Furthermore, the nuclear factor­κB (NF­κB) pathway was activated following cell treatment with CTX, and NF­κB p65 overexpression attenuated CTX cytotoxicity. In addition, the combined use of the specific NF­κB inhibitor caffeic acid phenethyl ester (CAPE) with CTX significantly enhanced CTX effect, both in vivo and in vitro. Similarly, the mRNA and protein expression of B­cell lymphoma-2 was decreased in AsPC­1 and BxPC­3 cells following treatment with CTX and CAPE, suggesting that NF­κB may serve a crucial role in CTX efficiency. In conclusion, results from our previous study indicated that CTX could potentially replace PTX in the treatment of PC, and the present study demonstrated that CTX combination with an NF­κB inhibitor may be considered as a potential therapeutic option for PC, which may improve the prognosis of patients with PC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ácidos Cafeicos/farmacologia , NF-kappa B/antagonistas & inibidores , Neoplasias Pancreáticas/tratamento farmacológico , Álcool Feniletílico/análogos & derivados , Taxoides/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Ácidos Cafeicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , NF-kappa B/metabolismo , Neoplasias Pancreáticas/patologia , Álcool Feniletílico/farmacologia , Álcool Feniletílico/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Taxoides/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Am J Cancer Res ; 10(1): 60-77, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32064153

RESUMO

As a member of epidermal growth factor receptor (EGFR) kinase substrate 8 (EPS8) family, the role of EPS8 like 3 protein (EPS8L3) has not been well studied in malignancies. However, EPS8 has been reported to be associated with prognosis and functions in several kinds of cancers. Hence, whether EPS8L3 plays similar roles in the tumorigenesis of human cancers, especially in hepatocellular carcinoma (HCC), is still needed to be further explored. In this study, we revealed that EPS8L3 was overexpressed in HCC tissues compared with adjacent non-tumor tissues, and was associated with a poor clinical prognosis. Both in vitro and in vivo experiments showed that EPS8L3 could promote the proliferative ability by downregulating p21/p27 expression, and promote the migratory and invasive abilities by upregulating matrix metalloproteinase-2 expression. Furthermore, we demonstrated that EPS8L3 could affect the activation of the EGFR-ERK pathway by modulating EGFR dimerization and internalization, which may not depend on the formation of EPS8L3-SOS1-ABI1 complex. Taken together, our study showed that EPS8L3 plays a pivotal role in the tumorigenesis and progression of HCC, and it might be a potential therapeutic target for HCC.

13.
Clin Transl Oncol ; 22(3): 392-400, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31264147

RESUMO

BACKGROUND: Intrahepatic cholangiocarcinoma (ICC) is one of the most devastating cancers of the gastrointestinal tract. It is crucial to determine the accurate prognostic factors and find new therapeutic strategies. Meanwhile, O6-methylguanine-DNA methyltransferase (MGMT) is associated with malignant tumor progression. Thus, further studies are needed to investigate whether MGMT plays a similar role in ICC. MATERIALS AND METHODS: Quantitative real-time PCR, western blot, and immunohistochemistry staining were used to detect the expression of MGMT in ICC tissues. The correlations between MGMT expression and clinicopathologic features were analyzed. The cell-proliferation assay and colony-formation assay were applied to evaluate proliferation ability, while methylation-specific PCR were used to detect the methylation status of the MGMT promoter CpG island in ICC tissues and cells. RESULTS: Our study found that the expression of MGMT was decreased in ICC tissues when compared with paired normal tissues. In addition, we demonstrated that MGMT expression was positively correlated with overall survival rates and tumor histological grade. Silencing of MGMT significantly promoted cell proliferation in ICC. Further research showed that silencing of MGMT induced cells to enter S phase by inhibiting p21, p27, and Cyclin E expression, ultimately promoting ICC proliferation. We also demonstrated that the MGMT promoter was highly methylated in ICC, and the levels of MGMT and p21 mRNA increased after DNA demethylation. In addition, the levels of MGMT and p21 protein were positively correlated in ICC tissues. CONCLUSION: MGMT may play a critical role in carcinogenesis and the development of ICC, and provides a new marker of clinical prognosis and target for ICC treatment.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Proliferação de Células/genética , Colangiocarcinoma/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Ductos Biliares Intra-Hepáticos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Ilhas de CpG , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Prognóstico , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/genética
14.
J Cancer ; 10(8): 1846-1854, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31205541

RESUMO

Cell division cycle associated 5 (CDCA5) is an important element for the interaction between cohesin and chromatin in interphase. It is abnormally expressed in many types of cancer and works as an indicator of poor prognosis, but little is known about its activity in hepatocellular carcinoma (HCC). In the present study, we found that the expression of CDCA5 was upregulated in HCC tissues compared to paracancerous tissues and had a negative correlation with patient survival. Cell proliferation and tumorigenesis were inhibited and cell apoptosis was induced with the knockdown of CDCA5, suggesting an oncogenic role of CDCA5 in liver cancer. Luciferase reporter assay and chromatin immunoprecipitation showed that CDCA5 was transcribed by E2F1. Furthermore, we confirmed that CDCA5 interrupted cell behavior via the AKT pathway. These findings demonstrated that CDCA5 plays an important role in HCC progression.

15.
Int J Cancer ; 145(2): 503-516, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30628729

RESUMO

The intermediate conductance calcium-activated potassium channel (KCa3.1) plays an important role in maintaining intracellular calcium homeostasis and is involved in the tumorigenesis of many human cancers. However, it is unknown whether KCa3.1 plays a role in the genesis of hepatocellular carcinoma (HCC), one of the most common malignant tumors worldwide with a very poor prognosis. In our study, we found that the expression of KCa3.1 was significantly elevated in poorly differentiated HCC tissues compared to adjacent noncancerous tissues. In vitro and in vivo experiments showed that KCa3.1 could promote cell proliferation, migration, and invasion of HCC. Mechanistically, KCa3.1 promoted cell cycle progression and migration and invasion of HCC cells by activating S-phase protein kinase 2 (SKP2) to trigger the degradation of p21 and p27 and targeting Reelin (RELN) to induce epithelial-mesenchymal transition (EMT), respectively. Taken together, our results demonstrate that KCa3.1 plays an important role in the genesis and progression of HCC, implying that it might be a promising therapeutic target in HCC.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/secundário , Proteínas Quinases Associadas a Fase S/genética , Transdução de Sinais , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Transplante de Neoplasias , Prognóstico , Proteína Reelina , Proteínas Quinases Associadas a Fase S/metabolismo , Ativação Transcricional , Regulação para Cima
16.
IEEE J Biomed Health Inform ; 23(3): 1215-1224, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29994412

RESUMO

Thyroid ultrasonography is a widely used clinical technique for nodule diagnosis in thyroid regions. However, it remains difficult to detect and recognize the nodules due to low contrast, high noise, and diverse appearance of nodules. In today's clinical practice, senior doctors could pinpoint nodules by analyzing global context features, local geometry structure, and intensity changes, which would require rich clinical experience accumulated from hundreds and thousands of nodule case studies. To alleviate doctors' tremendous labor in the diagnosis procedure, we advocate a machine learning approach to the detection and recognition tasks in this paper. In particular, we develop a multitask cascade convolution neural network (MC-CNN) framework to exploit the context information of thyroid nodules. It may be noted that our framework is built upon a large number of clinically confirmed thyroid ultrasound images with accurate and detailed ground truth labels. Other key advantages of our framework result from a multitask cascade architecture, two stages of carefully designed deep convolution networks in order to detect and recognize thyroid nodules in a pyramidal fashion, and capturing various intrinsic features in a global-to-local way. Within our framework, the potential regions of interest after initial detection are further fed to the spatial pyramid augmented CNNs to embed multiscale discriminative information for fine-grained thyroid recognition. Experimental results on 4309 clinical ultrasound images have indicated that our MC-CNN is accurate and effective for both thyroid nodules detection and recognition. For the correct diagnosis rate of malignant and benign thyroid nodules, its mean Average Precision (mAP) performance can achieve up to [Formula: see text] accuracy, which outperforms the common CNNs by [Formula: see text] on average. In addition, we conduct rigorous user studies to confirm that our MC-CNN outperforms experienced doctors, yet only consuming roughly [Formula: see text] ( 1/48) of doctors' examination time on average. Therefore, the accuracy and efficiency of our new method exhibit its great potential in clinical applications.


Assuntos
Interpretação de Imagem Assistida por Computador/métodos , Nódulo da Glândula Tireoide/diagnóstico por imagem , Algoritmos , Humanos , Redes Neurais de Computação , Glândula Tireoide/diagnóstico por imagem , Ultrassonografia
17.
BMC Microbiol ; 18(1): 87, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30119655

RESUMO

BACKGROUND: The pathogenesis of Helicobacter pylori (H. pylori) infection-induced duodenal ulcer remains to be elucidated. Duodenal mucosal bicarbonate secretion is the most important protective factor against acid-induced mucosal injury. We previously revealed that H. pylori infection downregulated the expression and functional activity of duodenal mucosal cystic fibrosis transmembrane conductance regulator (CFTR) and solute linked carrier 26 gene family A6 (SLC26A6) which are the two key duodenal mucosal epithelial cellular bicarbonate transporters to mediate duodenal bicarbonate secretion. In this study, we investigated the mechanism of H. pylori infection-induced duodenal CFTR and SLC26A6 expression downregulation. RESULTS: We found that H. pylori infection induced the increase of serum transforming growth factor ß (TGFß) level and duodenal mucosal TGFß expression and the decrease of duodenal mucosal CFTR and SLC26A6 expressions in C57 BL/6 mice. The results from the experiments of human duodenal epithelial cells (SCBN) showed that H. pylori increased TGFß production and decreased CFTR and SLC26A6 expressions in SCBN cells. TGFß inhibitor SB431542 reversed the H. pylori-induced CFTR and SLC26A6 expression decreases. The further results showed that TGFß directly decreased CFTR and SLC26A6 expressions in SCBN cells. TGFß induced the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and P38 MAPK inhibitor SB203580 reversed the TGFß-induced CFTR and SLC26A6 expression decreases. CONCLUSIONS: H. pylori infection downregulates duodenal epithelial cellular CFTR and SLC26A6 expressions through TGFß-mediated P38 MAPK signaling pathway, which contributes to further elucidating the pathogenesis of H. pylori-associated duodenal ulcer.


Assuntos
Antiporters/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulação para Baixo , Duodeno/metabolismo , Infecções por Helicobacter , Helicobacter pylori/patogenicidade , Transportadores de Sulfato/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Antiporters/genética , Bicarbonatos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Duodeno/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Infecções por Helicobacter/microbiologia , Imidazóis/antagonistas & inibidores , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/antagonistas & inibidores , Transportadores de Sulfato/genética , Fator de Crescimento Transformador beta/genética , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/genética
18.
Biochem Biophys Res Commun ; 499(3): 433-440, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29574153

RESUMO

Kelch-like family member 21 (KLHL21) is involved in cell mitosis and motility. Nevertheless, the clinical significance and biological function of KLHL21 in cholangiocarcinoma (CCA) are elusive. This is the first study to describe a pivotal role for KLHL21 in the progression of CCA. The expression of KLHL21 was elevated in CCA tissues compared with paired normal bile duct tissues. In addition, immunohistochemical and statistical analyses demonstrated that the expression of KLHL21 correlated inversely with tumor histological grade (p < 0.05) and the overall survival of patients (p < 0.01). In CCA cells, we found that the inhibition of KLHL21 significantly reduced proliferation, migration and invasion. Further results indicated that inhibition of KLHL21 triggered G0/G1 cell cycle arrest, leading to the increased expression of P21 and P27 and decreased expression of Cyclin E1, which eventually resulted in proliferation suppression in CCA cells. Furthermore, KLHL21 knockdown alleviated the activation of the Erk signaling pathway via decreasing the expression of phospho-Erk1/2. Our data demonstrated that KLHL21 plays an essential role in the tumorigenesis and progression of CCA, implying that it might serve as a potential therapeutic target for CCA treatment.


Assuntos
Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/antagonistas & inibidores , Movimento Celular , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patologia , Proteínas do Citoesqueleto/antagonistas & inibidores , Progressão da Doença , Sistema de Sinalização das MAP Quinases , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Colangiocarcinoma/enzimologia , Proteínas do Citoesqueleto/metabolismo , Ativação Enzimática , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Invasividade Neoplásica , Prognóstico , RNA Interferente Pequeno/metabolismo , Análise de Sobrevida
19.
Head Neck ; 40(4): 778-783, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29286180

RESUMO

BACKGROUND: The purpose of this study was to compare the diagnostic efficiency of a thyroid ultrasound computer-aided diagnosis (CAD) system with that of 1 radiologist. METHODS: This study retrospectively reviewed 342 surgically resected thyroid nodules from July 2013 to December 2013 at our center. The nodules were assessed on typical ultrasound images using the CAD system and reviewed by 1 experienced radiologist. The radiologist stratified the risk of malignancy using the Thyroid Imaging Reporting and Data Systems (TIRADS) and the American Thyroid Association (ATA) guidelines. RESULTS: The radiologist, using TI-RADS and ATA guidelines, performed better than the CAD system (P < .01). The sensitivity of the CAD system was similar to that of an experienced radiologist (P > .05; P < .01; and P > .05). However, we found that the CAD system had lower specificity (P < .01). CONCLUSION: The sensitivity of a thyroid ultrasound CAD system in differentiating nodules was similar to that of an experienced radiologist. However, the CAD system had lower specificity.


Assuntos
Diagnóstico por Computador/métodos , Biópsia Guiada por Imagem/métodos , Neoplasias da Glândula Tireoide/patologia , Nódulo da Glândula Tireoide/patologia , Ultrassonografia de Intervenção/métodos , Adulto , China , Estudos de Coortes , Feminino , Hospitais Universitários , Humanos , Masculino , Pessoa de Meia-Idade , Curva ROC , Radiologistas , Estudos Retrospectivos , Sensibilidade e Especificidade , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/cirurgia , Nódulo da Glândula Tireoide/diagnóstico por imagem , Nódulo da Glândula Tireoide/cirurgia , Tireoidectomia/métodos
20.
J Cancer ; 8(9): 1568-1578, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28775776

RESUMO

Background: Intrahepatic cholangiocarcinoma (ICC) is a high malignant tumor arising from the bile ducts in the liver with a poor prognosis. As current molecular targeted therapies and systemic chemotherapies had limited success in ICC, novel therapeutic targets are needed. In this study, we attempted to investigate the expression and the role of the intermediate conductance calcium-activated potassium channel (KCa3.1) in ICC. Methods: The expression levels of KCa3.1 channel were measured in 81 resected ICC tumor specimens and the clinicopathological significance of these levels were determined. KCa3.1 channel inhibitor and siRNA were used to study the role of KCa3.1 in proliferation, migration, and invasion of ICC cell lines. The effect of KCa3.1 channel blockade on tumor growth in vivo was also studied using xenograft model in nude mice. Results: The protein expression of KCa3.1 channel was upregulated in ICC tissues and was correlated with age, lymph node metastasis and TNM stage. And high KCa3.1 expression indicated a worse prognosis in ICC patients. Blocking KCa3.1 channel with a specific inhibitor TRAM-34 reduced the proliferation and invasion of ICC cells. Knockdown of KCa3.1 could achieve the same effects through decreasing NF-κB activation. Further in vivo studies demonstrated that KCa3.1 channel blockade suppressed ICC tumor growth. Conclusions: Our observations suggested KCa3.1 might be a promising novel therapeutic target in intrahepatic cholangiocarcinoma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA