Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Gastroenterology ; 153(1): 292-306.e2, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28390868

RESUMO

BACKGROUND & AIMS: Small nucleolar noncoding RNAs (snoRNAs) regulate function of ribosomes, and specific snoRNAs are dysregulated in some cancer cells. We investigated dysregulation of snoRNAs in pancreatic ductal adenocarcinoma (PDAC) cells. METHODS: We investigated snoRNA expression in PDAC cell lines by complementary DNA microarray and quantitative reverse transcription polymerase chain reaction. In PDAC (n = 133), intraductal papillary mucinous neoplasm (n = 16), mucinous cystic neoplasm-associated PDAC (n = 1), and non-tumor pancreas (n = 8) and liver (n = 3) tissues from subjects who underwent surgical resection, levels of snoRNA were measured by quantitative reverse transcription polymerase chain reaction and compared with clinicopathologic parameters and survival times determined by Kaplan-Meier analysis. To examine snoRNA function, PDAC cells were transfected with snoRNA-antisense oligonucleotides flanked with amido-bridged nucleic acids, or snoRNA-expression plasmids, and analyzed in proliferation, colony formation, spheroid formation, and invasion assays. To identify snoRNA-related factors, cells were analyzed by gene expression and proteomic profiling and immunoblot assays. Mice were given intrasplenic injections of MIA PaCa2- or Suit2-HLMC cells; tumor-bearing nude mice were then given 3 weekly injections of an antisense oligonucleotides against SNORA23, a H/ACA-box type snoRNA, and tumor growth and metastasis to liver, blood, and pancreas were analyzed. RESULTS: Levels of SNORA23 increased and accumulated at the nucleolus in highly metastatic MIA PaCa2- or Suit2-HLMC cells compared with their parental cells. We detected SNORA23 in human PDAC specimens but not in non-tumor pancreatic tissue. PDAC level of SNORA23 correlated with invasion grade and correlated inversely with disease-free survival time of patients. Expression of SNORA23 in PDAC cells increased their invasive activity and colony formation, and spheroid formation was inhibited by SNORA23 knockdown. In gene expression and proteomic profile analyses, we found SNORA23 to increase expression of spectrin repeat-containing nuclear envelope 2 (SYNE2) messenger RNA and protein. Knockdown of SYNE2 in PDAC cells reduced their invasive activities and anchor-independent survival. Administration of SNORA23 antisense oligonucleotides to mice slowed growth of xenograft tumors, tumor expression of SYNE2, tumor cell dissemination, and metastasis to liver. CONCLUSIONS: We found expression of the snoRNA SNORA23, which mediates sequence-specific pseudouridylation of ribosomal RNAs, to be increased in human PDAC tissues compared with non-tumor tissues, and levels to correlate with tumor invasion grade and patient survival time. SNORA23 increases expression of SYNE2, possibly through modulation of ribosome biogenesis, to promote PDAC cell survival and invasion, and growth and metastasis of xenograft tumors in mice.


Assuntos
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundário , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/secundário , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , RNA Nucleolar Pequeno/genética , Animais , Carcinoma Ductal Pancreático/química , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Proliferação de Células/genética , DNA Complementar/análise , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Carioferinas/genética , Masculino , Camundongos , Proteínas dos Microfilamentos/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Transplante de Neoplasias , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso/uso terapêutico , Pâncreas/química , Neoplasias Pancreáticas/química , Proteoma , RNA Mensageiro/metabolismo , RNA Nucleolar Pequeno/análise , RNA Nucleolar Pequeno/antagonistas & inibidores , RNA Nucleolar Pequeno/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Esferoides Celulares , Taxa de Sobrevida , Transfecção , Regulação para Cima
2.
Tumour Biol ; 36(2): 849-59, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25296734

RESUMO

Cyclophilin A (CypA) was shown to be upregulated in human cholangiocarcinoma (CCA) tissues. Suppression of intracellular CypA (inCypA) significantly reduces cell proliferation in vitro and tumor growth in nude mice. In the present study, the effect and potential mechanism of secreted CypA (sCypA) on cell proliferation of CCA cell lines were further investigated. CCA cells were treated with sCypA-containing conditioned media (CM) or with purified recombinant human CypA (rhCypA). Cell proliferation, cell cycle, ERK1/2, p38 MAPK, NF-κB, and STAT3 activities were examined by MTS assay, flow cytometry, and Western blot. sCypA was detected in CM from MMNK1 (an immortalized human cholangiocyte cell line) and six CCA cell lines. The sCypA levels corresponded to the inCypA levels indicating the intracellular origin of sCypA. Both sCypA-containing CM and rhCypA significantly increased proliferation of CCA cells. CD147 depletion by shRNA-knockdown or neutralizing with a CD147-monoclonal antibody significantly reduced sCypA-, and rhCypA-mediated cell proliferation. Upon rhCypA treatment, ERK1/2 was rapidly phosphorylated; whereas neutralizing CD147 inhibited ERK1/2 phosphorylation. Cell cycle analysis showed a significant increase in S phase and decrease in G1 population in rhCypA-treated cells. The expression levels of cyclin D1 and phosphorylated-retinoblastoma protein in the rhCypA-treated cells were increased compared with those in the non-treated control cells. p38 MAPK pathway was shown to be suppressed in siCypA-treated cells. In summary, CypA is secreted from CCA cells and enhances cell proliferation in an autocrine/paracrine manner, at least via direct binding with CD147, which may activate the ERK1/2 and p38 MAPK signaling pathways.


Assuntos
Basigina/biossíntese , Neoplasias dos Ductos Biliares/genética , Colangiocarcinoma/genética , Ciclofilina A/genética , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Animais , Basigina/genética , Neoplasias dos Ductos Biliares/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Colangiocarcinoma/patologia , Meios de Cultivo Condicionados , Ciclofilina A/biossíntese , Ciclofilina A/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
3.
PLoS One ; 19(6): e0306060, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38923999

RESUMO

Cholangiocarcinoma (CCA) is an aggressive cancer originating from bile duct epithelium, particularly prevalent in Asian countries with liver fluke infections. Current chemotherapy for CCA often fails due to drug resistance, necessitating novel anticancer agents. This study investigates the potential of 5'-deoxy-5'-methylthioadenosine (MTA), a naturally occurring nucleoside, against CCA. While MTA has shown promise against various cancers, its effects on CCA remain unexplored. We evaluated MTA's anticancer activity in CCA cell lines and drug-resistant sub-lines, assessing cell viability, migration, invasion, and apoptosis. The potential anticancer mechanisms of MTA were explored through proteomic analysis using LC-MS/MS and bioinformatic analysis. The results show a dose-dependent reduction in CCA cell viability, with enhanced effects on cancer cells compared to normal cells. Moreover, MTA inhibits growth, induces apoptosis, and suppresses cell migration and invasion. Additionally, MTA enhanced the anticancer effects of gemcitabine on drug-resistant CCA cells. Proteomics revealed the down-regulation of multiple proteins by MTA, affecting various molecular functions, biological processes, and cellular components. Network analysis highlighted MTA's role in inhibiting proteins related to mitochondrial function and energy derivation, crucial for cell growth and survival. Additionally, MTA suppressed proteins involved in cell morphology and cytoskeleton organization, important for cancer cell motility and metastasis. Six candidate genes, including ZNF860, KLC1, GRAMD1C, MAMSTR, TANC1, and TTC13, were selected from the top 10 most down-regulated proteins identified in the proteomics results and were subsequently verified through RT-qPCR. Further, KLC1 protein suppression by MTA treatment was confirmed through Western blotting. Additionally, based on TCGA data, KLC1 mRNA was found to be upregulated in the tissue of CCA patients compared to that of normal adjacent tissues. In summary, MTA shows promising anticancer potential against CCA by inhibiting growth, inducing apoptosis, and suppressing migration and invasion, while enhancing gemcitabine's effects. Proteomic analysis elucidates possible molecular mechanisms underlying MTA's anticancer activity, laying the groundwork for future research and development of MTA as a treatment for advanced CCA.


Assuntos
Apoptose , Neoplasias dos Ductos Biliares , Movimento Celular , Colangiocarcinoma , Desoxiadenosinas , Proteômica , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patologia , Humanos , Proteômica/métodos , Linhagem Celular Tumoral , Desoxiadenosinas/farmacologia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/metabolismo , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Tionucleosídeos/farmacologia , Antineoplásicos/farmacologia , Gencitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos
4.
BMC Cancer ; 13: 430, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-24053380

RESUMO

BACKGROUND: Thymosin ß10 (Tß10) expression is associated with malignant phenotypes in many cancers. However, the role and mechanisms of Tß10 in liver fluke-associated cholangiocarcinoma (CCA) are not fully understood. In this study, we investigated the expression of Tß10 in CCA tumor tissues and cell lines as well as molecular mechanisms of Tß10 in tumor metastasis of CCA cell lines. METHODS: Tß10 expression was determined by real time RT-PCR or immunocytochemistry. Tß10 silence or overexpression in CCA cells was achieved using gene delivery techniques. Cell migration was assessed using modified Boyden chamber and wound healing assay. The effect of silencing Tß10 on CCA tumor metastasis was determined in nude mice. Phosphorylation of ERK1/2 and the expression of EGR1, Snail and matrix metalloproteinases (MMPs) were studied. RESULTS: Ten pairs of CCA tissues (primary and metastatic tumors) and 5 CCA cell lines were studied. With real time RT-PCR and immunostaining analysis, Tß10 was highly expressed in primary tumors of CCA; while it was relatively low in the metastatic tumors. Five CCA cell lines showed differential expression levels of Tß10. Silence of Tß10 significantly increased cell migration, invasion and wound healing of CCA cells in vitro; reversely, overexpression of Tß10 reduced cell migration compared with control cells (P<0.05). In addition, silence of Tß10 in CCA cells increased liver metastasis in a nude mouse model of CCA implantation into the spleen. Furthermore, silence of Tß10 activated ERK1/2 and increased the expression of Snail and MMPs in CCA cell lines. Ras-GTPase inhibitor, FPT inhibitor III, effectively blocked Tß10 silence-associated ERK1/2 activation, Snail expression and cell migration. CONCLUSIONS: Low expression of Tß10 is associated with metastatic phenotype of CCA in vitro and in vivo, which may be mediated by the activation of Ras, ERK1/2 and upregulation of Snail and MMPs. This study suggests a new molecular pathway of CCA pathogenesis and a novel strategy to treat or prevent CCA metastasis.


Assuntos
Movimento Celular/genética , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Timosina/genética , Animais , Linhagem Celular Tumoral , Colangiocarcinoma/etiologia , Colangiocarcinoma/metabolismo , Modelos Animais de Doenças , Fasciola hepatica , Fasciolíase/complicações , Expressão Gênica , Inativação Gênica , Humanos , Camundongos , Camundongos Nus , Metástase Neoplásica , Interferência de RNA , Timosina/metabolismo
5.
Expert Rev Anticancer Ther ; 23(5): 517-530, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37052887

RESUMO

INTRODUCTION: Cholangiocarcinoma (CCA) is an aggressive cancer arising from any part of the biliary system. Effective treatment of CCA remains limited, resulting in the poor overall prognosis of patients. The effective prognostic biomarkers for CCA remain lacking, and most are at the research level. AREAS COVERED: The incidences of CCAs, classification, genetic and molecular characteristics, and distinct clinical outcomes in each subtype are introduced. The prognostic markers currently used in clinical practice are reviewed. Studies of biomarkers in defining the aggressiveness of CCA, identifying patients with a potential tumor recurrence, and predicting the survival time, are reviewed. Emerging biomarkers discovered from advanced high throughput technology over the past 5 years are updated and summarized. Finally, in-depth and critical revision on the prognostic biomarkers for CCA reported from various sources of specimens, e.g. tissues, blood, bile, etc. are discussed. Conclusion: Many prognostic biomarkers for CCA have been proposed and hold promising clinical value. However, these markers are rarely used in the real clinical world due to several factors. Understanding the roles and importance of these prognostic markers may fundamentally impact the therapeutic management of CCA, and hopefully, improve the development of custom and patient-directed therapies for CCA.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Humanos , Prognóstico , Neoplasias dos Ductos Biliares/diagnóstico , Neoplasias dos Ductos Biliares/terapia , Neoplasias dos Ductos Biliares/genética , Recidiva Local de Neoplasia/patologia , Colangiocarcinoma/diagnóstico , Colangiocarcinoma/terapia , Ductos Biliares Intra-Hepáticos , Biomarcadores , Biomarcadores Tumorais/genética
6.
Artigo em Inglês | MEDLINE | ID: mdl-37294416

RESUMO

Cholangiocarcinoma (CCA), a bile duct cancer with a high mortality rate, has a poor prognosis due to its highly invasive and drug-resistant phenotypes. More effective and selective therapies are urgently needed. Bacteriocins are broad-spectrum antimicrobial peptides/proteins produced by bacterial strains to compete with other bacteria. Recent studies have reported that bacteriocins exhibit anticancer properties against various cancer cell lines with minimal toxicity toward normal cells. In this study, two types of recombinant bacteriocins, rhamnosin from probiotic Lacticaseibacillus rhamnosus and lysostaphin from Staphylococcus simulans, were highly produced in Escherichia coli and subsequently purified via immobilized-Ni2+ affinity chromatography. When their anticancer activity was investigated against CCA cell lines, both rhamnosin and lysostaphin were found capable of inhibiting the growth of CCA cell lines in a dose-dependent fashion but were less toxic toward a normal cholangiocyte cell line. Rhamnosin and lysostaphin as single treatments could suppress the growth of gemcitabine-resistant cell lines to the same extent as or more than they suppressed the parental counterparts. A combination of both bacteriocins more strongly inhibited growth and enhanced cell apoptosis in both parental and gemcitabine-resistant cells partly through the increased expression of the proapoptotic genes BAX, and caspase-3, -8, and -9. In conclusion, this is the first report to demonstrate an anticancer property of rhamnosin and lysostaphin. Using these bacteriocins as single agents or in combination would be effective against drug-resistant CCA.

7.
Dev Comp Immunol ; 135: 104459, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35660488

RESUMO

Serine proteases are proteolytic enzymes that exhibit biological roles in many biological systems. Previously, a Vibrio parahaemolyticus serine protease was reported to be a virulence factor. Here, the serine protease gene of V. parahaemolyticus was investigated as a DNA vaccine against V. parahaemolyticus in Litopenaeus vannamei. The serine protease gene was mutated to replace the conserved residues His82, Asp131 and Ser231 with Gly, Asp and Pro, respectively. Then, a pcDNA3.1 vector to express mutVpSP (mutant serine protease) was constructed for in vitro and in vivo DNA vaccine investigation. In vivo mutVpSP transcriptional analysis revealed expression in various immunized white shrimp tissues, such as hemocytes, hepatopancreas, stomach, intestine, gills, and muscle. The efficiency of prevention of V. parahaemolyticus infection was investigated in vaccinated shrimp, and the lowest cumulative mortality percentage was 30%, while the control shrimp had a 60% cumulative mortality rate. The immune system was stimulated in shrimp vaccinated with the DNA vaccine. The mRNA expression of the shrimp immune-responsive genes phenoloxidase, peroxinectin and C-type lectin was significantly upregulated. Additionally, the humoral and cellular immune responses, including the PO, phagocytic, and encapsulation activities and nodule formation, were elevated. These results suggested that the serine protease could be a V. parahaemolyticus virulence determinant and that this DNA vaccine could be applied as an effective vaccine candidate for control of acute hepatopancreatic necrosis disease syndrome (AHPND) in shrimp.


Assuntos
Penaeidae , Serina Proteases , Vacinas de DNA , Vibrioses , Vibrio parahaemolyticus , Animais , Imunidade Inata , Penaeidae/imunologia , Penaeidae/virologia , Serina , Serina Proteases/genética , Vibrioses/prevenção & controle , Vibrioses/veterinária
8.
Asian Pac J Cancer Prev ; 23(2): 715-721, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35225485

RESUMO

OBJECTIVE: Annexin A1 (ANXA1) is a calcium-dependent phospholipid-binding protein which contributes to proliferation, cancer progression and metastasis. Overexpression of ANXA1 is closely associated with metastasis in numerous types of cancer. Cholangiocarcinoma (CCA) is a bile-duct cancer which has high rates of metastasis. Previously, we demonstrated up-regulation of ANXA1 in a highly metastatic CCA cell line (KKU-213AL5). Here, we investigated the functions of ANXA1 in the progression of CCA cell lines and evaluated its clinical impacts in human CCA tissues.  Methods: Effects of ANXA1 on metastatic potential of CCA cell lines were evaluated using cell-proliferation, clonogenic, migration and invasion assays. The expression of ANXA1 in 44 intrahepatic human CCA tissues was investigated using immunohistochemistry (IHC). The association of ANXA1 with clinicopathological features of CCA patients was analyzed. RESULTS: Silencing of ANXA1 expression using siRNA significantly decreased cell proliferation, colony formation, cell migration and invasion in the KKU-213AL5 cell line. IHC results showed low expression of ANXA1 in normal bile ducts in the non-tumor area. In contrast, high expression of ANXA1 in human CCA tissues was associated with advanced tumor stage, tumor size and presence of lymph-node metastasis. CONCLUSION: These findings strongly imply that ANXA1 contributes to the progression of CCA. ANXA1 can serve as a potential prognostic marker for CCA. Ablation of ANXA1 action may be an alternative strategy to prevent metastasis of CCA.


Assuntos
Anexina A1/genética , Neoplasias dos Ductos Biliares/genética , Colangiocarcinoma/genética , Metástase Neoplásica/genética , Ductos Biliares/metabolismo , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Humanos , Imuno-Histoquímica , Metástase Linfática/genética , Invasividade Neoplásica/genética , Prognóstico
9.
Asian Pac J Cancer Prev ; 23(10): 3379-3386, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36308362

RESUMO

BACKGROUND: Berberine (BBR), a natural isoquinoline alkaloid, possesses diverse pharmacological properties and anti-cancer effects that have been demonstrated in many in vitro and in vivo studies. In this study, the inhibitory effects and molecular mechanism of low dose BBR on EMT-induced cell migration, and invasion capability of cholangiocarcinoma (CCA) cell lines were demonstrated. METHODS: The commercially available BBR chloride powder with purity ≥ 95% was used in this study. Effects of BBR on cell growth of two human CCA cell lines, KKU-213A and KKU-213B were measured using MTT assay. The progressive phenotypes-cell adhesion, migration, and invasion were evaluated using cell adhesion, wound healing, and Boyden chamber assays. Molecular docking analysis was performed to assess the possible binding mode of BBR against EGFR, Erk, STAT3 and Akt. The effects of BBR on the activations of EGF/EGFR and its downstream effectors were demonstrated using Western blotting. RESULTS: BBR inhibited growth of CCA cells in a dose dependent manner. At sub-cytotoxic dose, BBR significantly inhibited cell adhesion, migration, invasion and decreased expression of vimentin, slug, and VEGFA of both CCA cell lines. Molecular docking suggested the simultaneous inhibitory activity of BBR on EGFR, Erk, STAT3 and Akt. The Western blot analyses revealed that upon the EGF/EGFR activation, BBR considerably attenuated the activations of EGFR, Erk, STAT3 and Akt. CONCLUSION: Low dose of BBR suppresses EMT and thus aggressiveness of CCA cells, in part by its multi-kinase inhibitor property on EGFR and its downstream pathways.  BBR might be beneficial for therapy of human CCA.


Assuntos
Antineoplásicos , Berberina , Neoplasias dos Ductos Biliares , Colangiocarcinoma , Humanos , Neoplasias dos Ductos Biliares/patologia , Berberina/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/uso terapêutico , Simulação de Acoplamento Molecular , Linhagem Celular Tumoral , Colangiocarcinoma/patologia , Movimento Celular , Proliferação de Células , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ductos Biliares Intra-Hepáticos/patologia , Receptores ErbB/metabolismo
10.
Mol Cancer ; 10: 102, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21871105

RESUMO

BACKGROUND: Cyclophilin A (CypA) expression is associated with malignant phenotypes in many cancers. However, the role and mechanisms of CypA in liver fluke-associated cholangiocarcinoma (CCA) are not presently known. In this study, we investigated the expression of CypA in CCA tumor tissues and CCA cell lines as well as regulation mechanisms of CypA in tumor growth using CCA cell lines. METHODS: CypA expression was determined by real time RT-PCR, Western blot or immunohistochemistry. CypA silence or overexpression in CCA cells was achieved using gene delivery techniques. Cell proliferation was assessed using MTS assay or Ki-67 staining. The effect of silencing CypA on CCA tumor growth was determined in nude mice. The effect of CypA knockdown on ERK1/2 activation was assessed by Western blot. RESULTS: CypA was upregulated in 68% of CCA tumor tissues. Silencing CypA significantly suppressed cell proliferation in several CCA cell lines. Likewise, inhibition of CypA peptidyl-prolyl cis-trans isomerase (PPIase) activity using cyclosporin A (CsA) decreased cell proliferation. In contrast, overexpression of CypA resulted in 30% to 35% increases in proliferation of CCA cell lines. Interestingly, neither silence nor overexpression of CypA affected cell proliferation of a non-tumor human cholangiocyte cell line, MMNK1. Suppression of CypA expression attenuated ERK1/2 activity in CCA M139 cells by using both transient and stable knockdown methods. In the in vivo study, there was a 43% reduction in weight of tumors derived from CypA-silenced CCA cell lines compared with control vector CCA tumors in mice; these tumors with stable CypA silencing showed a reduced cell proliferation. CONCLUSIONS: CypA is upregulated in majority of CCA patients' tissues and confers a significant growth advantage in CCA cells. Suppression of CypA expression decreases proliferation of CCA cell lines in vitro and reduces tumor growth in the nude mouse model. Inhibition of CypA activity also reduces CCA cell proliferation. The ERK1/2 pathway may be involved in the CypA-mediated CCA cell proliferation. Thus, CypA may represent an important new therapeutic target for liver fluke-associated CCA.


Assuntos
Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos/patologia , Proliferação de Células , Colangiocarcinoma/genética , Ciclofilina A/fisiologia , Fasciola hepatica/fisiologia , Animais , Neoplasias dos Ductos Biliares/etiologia , Neoplasias dos Ductos Biliares/parasitologia , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/parasitologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colangiocarcinoma/etiologia , Colangiocarcinoma/parasitologia , Colangiocarcinoma/patologia , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/genética , Ciclofilina A/metabolismo , Fasciolíase/complicações , Fasciolíase/genética , Fasciolíase/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Nus , RNA Interferente Pequeno/farmacologia , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Life Sci ; 286: 120072, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34688691

RESUMO

AIMS: 5-Fluorouracil (5-FU), a thymidylate synthase (TS) inhibitor, has been used as the first-line chemotherapeutic drug for cholangiocarcinoma (CCA). The side effects and drug resistance have developed the limits of the clinical application of 5-FU in CCA treatment. Upregulation of Forkhead box M1 (FOXM1) and TS were shown to play a significant role in 5-FU resistance. In this study, the effect of Siomycin A (SioA), a FOXM1 inhibitor, on enhancing 5-FU cytotoxicity and reversing 5-FU resistance in CCA cell lines were demonstrated. MAIN METHODS: Human CCA cell lines, KKU-100 and KKU-213A were used. Cell viability was determined using MTT assay. Expression of FOXM1 and TS proteins were determined using Western blotting. FOXM1 mRNA expression was quantitated using real-time PCR. The combination and dose reduction (DRI) were analyzed according to the Chou and Talalay method. KEY FINDING: Single drug treatment of 5-FU and SioA effectively inhibited CCA cell growth in dose and time dependent fashions. The two CCA cell lines had different responses to 5-FU but exhibited similar sensitivity to SioA. FOXM1 and TS expression were increased in the 5-FU treated cells but were suppressed in the SioA treated cells. A direct binding of SioA, to TS and 5,10-methylene-tetrahydrofolate as an inactive ternary complex was simulated. The combined treatment of 5-FU with SioA showed a synergistic effect with a high DRI and restored 5-FU sensitivity in the 5-FU resistant cells. SIGNIFICANCE: Targeting FOXM1 using SioA in combination with 5-FU might be a strategy to overcome the 5-FU resistance in CCA.


Assuntos
Colangiocarcinoma/tratamento farmacológico , Peptídeos/farmacologia , Timidilato Sintase/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colangiocarcinoma/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fluoruracila/farmacologia , Proteína Forkhead Box M1/antagonistas & inibidores , Proteína Forkhead Box M1/metabolismo , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Peptídeos/metabolismo , Timidilato Sintase/fisiologia
12.
Cell Oncol (Dordr) ; 43(2): 211-222, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31729681

RESUMO

PURPOSE: Cholangiocarcinoma (CCA) is an aggressive type of cancer. The major obstacles for treatment are its late presentation and the occurrence metastases. Targeting the metastatic process may serve as a treatment option. CD147 is a membrane protein that promotes CCA metastasis. High lactate levels in CCA are predicted to result from lactate dehydrogenase A expression and sensitivity to monocarboxylate transporter (MCT) inhibitors. An involvement of CD147 in MCT maturation has been reported, but the exact role of MCT in CCA is not clear. Here, we aimed to assess the mechanism of CD147-promoted CCA progression through MCT regulation. METHODS: The expression levels of CD147 and MCT-1/4 in human CCA tissues were determined by immunohistochemistry. Two CD147 knockout (CD147 KO) CCA cell (KKU-213) clones were established using the CRISPR/Cas9 system. Cell migration and invasion were determined using a Boyden chamber assay. Temporal protein levels were modified by siRNA, specific inhibitors and/or activators. The expression of target proteins was determined using Western blot analyses. RESULTS: CD147 and MCT-1/4 were found to be overexpressed in CCA tissues compared to normal bile duct tissues. In addition, we found that CD147 knockdown significantly alleviated CCA cell migration and invasion, concomitant with decreased pAkt, pFoxO3, pNF-κB (pp65) and MCT-1/4 levels. Conversely, we found that FoxO3 knockdown led to recovered migration/invasion abilities and increased pp65 and MCT-1/4 expression levels. The involvement of Akt in the regulation of MCT-1/4 expression through CD147 was established by inhibition and activation of Akt phosphorylation. CONCLUSION: Our data indicate that CD147 promotes the malignant progression of CCA cells by activating the Akt-FoxO3-NF-κB-MCT-1/4 axis. As such, CD147 may serve as a possible target for advanced CCA treatment.


Assuntos
Basigina/metabolismo , Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas/metabolismo , Basigina/genética , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/genética , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Transportadores de Ácidos Monocarboxílicos/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Invasividade Neoplásica , Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Transdução de Sinais/genética
13.
Med Sci Monit ; 15(11): RA221-32, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19865066

RESUMO

Cyclophilin A (CypA) is a cytosolic binding protein of the immunosuppressive drug cyclosporin A. CypA has an activity of peptidylprolyl cis-trans isomerase, which may play important roles in protein folding, trafficking, assembly, immune-modulator and cell signaling. Secreted form of CypA can act as growth factors in several cell types. On the other hand, CypA also involves in pathogenesis in several diseases including viral infection, cardiovascular disease and cancer. For example, CypA protein displays an unusually high expression in several cancer types and correlates with poor outcome of the patients. In this review, we have focused on the recently available clinical data of altered CypA expression in various tumor tissues and cell lines and discussed contributions and potential molecular mechanisms of CypA to tumor progression and clinical association. CypA could promote cancer cell proliferation, anti-apoptosis, cell migration/invasion and drug resistant in various cancer cell types. The mechanism by which CypA contributes to cancer progression appears to involve a complex interplay of proteins and pathways including membrane receptor for CypA, CD147, and CypA binding partners inside the cells. CypA could be used as a molecular target for cancer therapy.


Assuntos
Ciclofilina A/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Ciclofilina A/genética , Humanos , Neoplasias/genética , Neoplasias/patologia
14.
Mol Ther Nucleic Acids ; 9: 170-181, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29246296

RESUMO

Y-box binding protein-1 (YB-1), involved in cancer progression and chemoradiation resistance, is overexpressed in not only cancer cells but also tumor blood vessels. In this study, we investigated the potential value of amido-bridged nucleic acid (AmNA)-modified antisense oligonucleotides (ASOs) targeting YB-1 (YB-1 ASOA) as an antiangiogenic cancer therapy. YB-1 ASOA was superior to natural DNA-based ASO or locked nucleic acid (LNA)-modified YB-1 ASO in both knockdown efficiency and safety, the latter assessed by liver function. YB-1 ASOA administered i.v. significantly inhibited YB-1 expression in CD31-positive angiogenic endothelial cells, but not in cancer cells, in the tumors. With regard to the mechanism of its antiangiogenic effects, YB-1 ASOA downregulated both Bcl-xL/VEGFR2 and Bcl-xL/Tie signal axes, which are key regulators of angiogenesis, and induced apoptosis in vascular endothelial cells. In the xenograft tumor model that had low sensitivity to anti-VEGF antibody, YB-1 ASOA significantly suppressed tumor growth; not only VEGFR2 but also Tie2 expression was decreased in tumor vessels. In conclusion, YB-1/Bcl-xL/VEGFR2 and YB-1/Bcl-xL/Tie signal axes play pivotal roles in tumor angiogenesis, and YB-1 ASOA may be feasible as an antiangiogenic therapy for solid tumors.

15.
Anticancer Res ; 36(11): 5965-5973, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27793922

RESUMO

BACKGROUND/AIM: The treatment of cholangiocarcinoma (CCA) is still ineffective and the search for a novel treatment is needed. In this study, eight novel mono-triazole glycosides (W1-W8) were synthesized and tested for their anticancer activities in CCA cell lines. MATERIALS AND METHODS: The anti-proliferation effect and the underlying mechanisms of the triazole glycosides were explored. Viable cells were determined using the MTT test. RESULTS: Among glycosides tested, W4 and W5 exhibited the most potent anticancer activity in a dose- and time-dependent fashion. Flow cytometry and wstern blot analysis revealed that W4 and W5 induced G0/G1 phase cell-cycle arrest through down-regulation of cyclin D1, cyclin E and induction of cyclin-dependent kinase inhibitors, p27 and p21 protein expression. Annexin V/propidium iodide (PI) staining demonstrated that W4 and W5 also induced apoptotic cells in a dose-dependent manner via caspase signaling cascade. CONCLUSION: Together, these findings imply that the novel synthetic glycosides might be a promising anticancer agent for CCA.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Colangiocarcinoma/patologia , Glicosídeos/farmacologia , Triazóis/farmacologia , Linhagem Celular Tumoral , Glicosídeos/química , Humanos , Triazóis/química
16.
Oncol Rep ; 36(3): 1435-46, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27461717

RESUMO

Cholangiocarcinoma (CCA) is a highly metastatic tumor, and the lung is a common site of metastasis. A greater understanding of the biology of metastases is needed to improve treatment outcomes. Herein, a highly metastatic human CCA subline, KKU-213L5 from an original cell line, KKU-213 that has marginally metastatic ability, was established and characterized. KKU-213L5 was selected in vivo through the fifth serial passage of pulmonary metastasized tissues via tail-vein injection in NOD/scid/Jak3 mice. The metastatic abilities of the KKU-213L5 cells were compared with the parental line in vitro and in vivo. The expression profile of this metastatic cell line was determined using real-time PCR. KKU-213L5 cells were found to possess higher metastatic phenotypes, i.e., growth rates, stem cell surface markers (CD133), migration and invasion characteristics when compared with the parental cells. Compared to the KKU-213 cells, KKU-213L5 cells formed larger tumors in subcutaneous xenografted mice and had a >10-fold increase in lung metastases in the tail-vein injected metastatic mouse model. Mice injected intravenously with KKU-213L5 cells had a significantly shorter survival. Analysis of the expressed genes related to progression of cancer revealed significant upregulation of anterior gradient protein-2 (AGR2) and suppression of KiSS-1 in the KKU-213L5 cells. The association of these two genes with metastasis was affirmed in CCA patient tissues since increased AGR2 expression and decreased KiSS-1 expression were found in higher stage patient tumors. In conclusion, a highly metastatic human CCA cell line was established and characterized. It is plausible that the differential expression between the parental KKU-213 and highly metastatic KKU-213L5 cells may be beneficial to classify novel genes associated with metastasis. The KKU-213L5 cell line should serve as a valued device for discovering the molecular mechanisms of CCA metastasis and enabling the search for an effective therapy for the unmet clinical need in CCA.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/patologia , Metástase Neoplásica/patologia , Animais , Neoplasias dos Ductos Biliares/metabolismo , Linhagem Celular Tumoral , Colangiocarcinoma/metabolismo , Humanos , Kisspeptinas/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mucoproteínas , Proteínas Oncogênicas , Proteínas/metabolismo , Regulação para Cima/fisiologia
17.
Asian Pac J Cancer Prev ; 13 Suppl: 71-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23480768

RESUMO

Claudin-4 (CLDN4) is a tight junction protein that forms apical junctional complexes in epithelial and endothelial cellular sheets. Acting as a barrier and control of permeability are the general functions of tight junction proteins contributing to tissue homeostasis, paracellular ion flux, and cell-cell contact. In this study, we immunohistochemically examined CLDN4 expression in liver fluke-associated cholangiocarcinomas (CCAs) with tissue microarrays. Regardless of the histological type and gross type of cancer, high expression of CLDN4 was noted in precancerous hyperplastic/dysplastic biliary epithelia and CCA. To investigate functional roles of CLDN4 in cancer progression, the effects of CLDN4 suppression by siRNA on cell proliferation, migration and invasion were investigated in two CCA cell lines, KKU-M139 and KKU-M213. Suppression of CLDN4 expression did not alter cell proliferation but caused significant reduction of cell migration and invasion by both CCA cell lines. Our results suggest that over-expressed CLDN4 may promote CCA expansion and metastasis.


Assuntos
Colangiocarcinoma , Claudina-4 , Animais , Neoplasias dos Ductos Biliares , Ductos Biliares Intra-Hepáticos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA