Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Ecotoxicol Environ Saf ; 281: 116618, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38944011

RESUMEN

BACKGROUND: Gastric cancer is a leading cause of cancer-related deaths influenced by both genetic and environmental factors. Triphenyl phosphate (TPP) is a prevalent flame retardant, but its health implications remain to be thoroughly understood. OBJECTIVE: To explore the link between TPP exposure and gastric cancer by examining gene expression patterns and developing a predictive model. METHODS: Gene expression data were sourced from The Cancer Genome Atlas (TCGA) and the Comparative Toxicogenomics Database (CTD). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were employed for analysis. Single-sample Gene Set Enrichment Analysis (ssGSEA) was used to obtain phosphate flame retardant-related scores. A predictive model was constructed through differential analysis, univariate COX regression, and LASSO regression. Molecular docking was performed to assess protein interactions with TPP. RESULTS: ssGSEA identified scores related to phosphate flame retardants in gastric cancer, which had a strong association with immune-related traits. Several genes associated with TPP were identified and used to develop a prognostic model that has clinical significance. Molecular docking showed a high binding affinity of TPP with MTTP, a gene related to lipid metabolism. Pathway analysis indicated that TPP exposure contributes to gastric cancer through lipid metabolic processes. CONCLUSION: The study establishes a potential correlation between TPP exposure and gastric cancer onset, pinpointing key genes and pathways involved. This underscores the significance of environmental factors in gastric cancer research and presents a potential diagnostic tool for clinical application.

2.
Cell Biol Int ; 2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38825780

RESUMEN

Ferroptosis is a novel form of programmed cell death and is considered to be a druggable target for colorectal cancer (CRC) therapy. However, the role of ferroptosis in CRC and its underlying mechanism are not fully understood. In the present study we found that a protein enriched in the Golgi apparatus, Golgi phosphoprotein 3 (GOLPH3), was overexpressed in human CRC tissue and in several CRC cell lines. The expression of GOLPH3 was significantly correlated with the expression of ferroptosis-related genes in CRC. The overexpression of GOLPH3 in Erastin-induced Caco-2 CRC cells reduced ferroptotic phenotypes, whereas the knockdown of GOLPH3 potentiated ferroptosis in HT-29 CRC cells. GOLPH3 induced the expression of prohibitin-1 (PHB1) and prohibitin-2 (PHB2), which also inhibited ferroptosis in Erastin-treated CRC cells. Moreover, GOLPH3 interacted with PHB2 and nuclear factor erythroid 2-related factor 2 (NRF2) in Caco-2 cells. These observations indicate that GOLPH3 is a negative regulator of ferroptosis in CRC cells. GOLPH3 protects these cells from ferroptosis by inducing the expression of PHB1 and PHB2, and by interacting with PHB2 and NRF2.

3.
Front Pharmacol ; 15: 1337883, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38828452

RESUMEN

Background: The interaction between environmental endocrine-disrupting chemicals, such as Bisphenol A (BPA), and their influence on cancer progression, particularly regarding the GOLPH3 gene in colorectal cancer, remains unclear. Methods: We performed an integrated analysis of transcriptional profiling, clinical data, and bioinformatics analyses utilizing data from the Comparative Toxicogenomics Database and The Cancer Genome Atlas. The study employed ClueGO, Gene Set Enrichment Analysis, and Gene Set Variation Analysis for functional enrichment analysis, alongside experimental assays to examine the effects of BPA exposure on colorectal cancer cell lines, focusing on GOLPH3 expression and its implications for cancer progression. Results: Our findings demonstrated that BPA exposure significantly promoted the progression of colorectal cancer by upregulating GOLPH3, which in turn enhanced the malignant phenotype of colorectal cancer cells. Comparative analysis revealed elevated GOLPH3 protein levels in cancerous tissues versus normal tissues, with single-cell analysis indicating widespread GOLPH3 presence across various cell types in the cancer microenvironment. GOLPH3 was also associated with multiple carcinogenic pathways, including the G2M checkpoint. Furthermore, our investigation into the colorectal cancer microenvironment and genomic mutation signature underscored the oncogenic potential of GOLPH3, exacerbated by BPA exposure. Conclusion: This study provides novel insights into the complex interactions between BPA exposure and GOLPH3 in the context of colorectal cancer, emphasizing the need for heightened awareness and measures to mitigate BPA exposure risks. Our findings advocate for further research to validate these observations in clinical and epidemiological settings and explore potential therapeutic targets within these pathways.

4.
Biochem Pharmacol ; 225: 116274, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735445

RESUMEN

GOLPH3 has been identified as an oncoprotein, playing a crucial role on progression and chemoresistancein of colon adenocarcinoma (COAD). However, it is still unclear the regulation of GOLPH3 expression at protein level. We discovered ubiquitin-specific proteases 6 (USP6) directly regulated the deubiquitination of the GOLPH3 protein and enhanced its stability in COAD. Overexpression of USP6 promoted COAD cell viability, inhibited apoptosis, and accelerated the growth of transplanted tumors growth in vitro and in vivo by deubiquitinating GOLPH3. Additionally, circCYFIP2 showed high expression levels in DDP-resistant colon cancer cells, promoting the cell proliferation. Mechanically, circCYFIP2 binds to both GOLPH3 protein and USP6, strengthening the interaction between GOLPH3 and USP6, and consequently induced DDP resistance in vitro and in vivo. In conclusion, USP6 operates as a deubiquitinase, targeting the GOLPH3 protein in COAD and enhancing its stability. Meanwhile, circCYFIP2 is crucial for the deubiquitination of GOLPH3 protein mediated by USP6 and acts as a scaffold to confer platinum resistance. The discovery of circCYFIP2/USP6/GOLPH3 pathway offers a potential target for overcoming chemoresistance in COAD.


Asunto(s)
Neoplasias del Colon , Resistencia a Antineoplásicos , Proteínas de la Membrana , Ubiquitina Tiolesterasa , Ubiquitinación , Animales , Humanos , Masculino , Ratones , Antineoplásicos/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Neoplasias del Colon/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitinación/efectos de los fármacos
5.
Discov Oncol ; 15(1): 193, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38806777

RESUMEN

BACKGROUND: 5-fluorouracil (5-FU) is conventionally used in chemotherapy for colon adenocarcinomas. Acquired resistance of 5-FU remains a clinical challenge in colon cancer, and efforts to develop targeted agents to reduce resistance have not yielded success. Protosappanin B (PSB), the main component of Lignum Sappan extract, is known to exhibit anti-tumor effects. However, whether and how PSB could improve 5-FU resistance in colon cancer have not yet been established. In this study, we aimed to explore the effects and underlying mechanisms of PSB in 5-FU-induced chemoresistance in colon adenocarcinoma. METHODS: Forty-seven paired colon cancer tissue samples from patients who received 5-FU chemotherapy were collected as clinical samples. Two 5-FU resistant colon cancer cell lines were established for in vitro experiments. Reverse transcription-quantitative PCR (RT-qPCR) was performed to determine the mRNA and microRNA (miRNA) expression levels in colon adenocarcinoma tissues and cell lines. Cell Counting Kit-8 (CCK-8) and flow cytometry assays were performed to evaluate cell proliferation and apoptosis, respectively. RESULTS: LINC00612 was highly expressed in colon adenocarcinoma samples and 5-FU resistant colon cancer cells. LINC00612 knockdown enhances 5-FU chemosensitivity in 5-FU resistant cells. Notably, PSB treatment attenuated LINC00612 expression in 5-FU resistant colon adenocarcinoma cells. Moreover, PSB treatment reversed the increase in LINC00612-induced 5-FU resistance. Mechanistically, LINC00612 specifically bound to miR-590-3p, which promoted 5-FU resistance in colon adenocarcinoma cells and attenuated the inhibitory effect of LINC00612 on GOLPH3 expression. CONCLUSION: PSB attenuates 5-FU chemoresistance in colon adenocarcinoma by regulating the LINC00612/miRNA-590-3p/GOLPH3 axis.

6.
BMC Cancer ; 24(1): 175, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38317072

RESUMEN

BACKGROUND: Targeted drugs are the main methods of RCC treatment. However, drug resistance is common in RCC patients, in-depth study of the drug-resistant mechanism is essential. METHODS: We constructed sunitinib resistant and Twist overexpressed A498 cells, and studied its mechanisms in vitro and in vivo. RESULTS: In cell research, we found that either sunitinib resistance or Twist overexpression can activate Wnt/ß-catenin and EMT signaling pathway, and the sunitinib resistance may work through ß-catenin/TWIST/TCF4 trimer. In zebrafish research, we confirmed the similarity of Twist overexpression and sunitinib resistance, and the promoting effect of Twist overexpression on drug resistance. CONCLUSIONS: Sunitinib resistance and Twist overexpression can activate Wnt/ß-catenin signaling pathway and EMT to promote the growth and metastasis of RCC cells.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Animales , Humanos , Sunitinib/farmacología , Sunitinib/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo , Pez Cebra/metabolismo , Línea Celular Tumoral , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Transición Epitelial-Mesenquimal/genética , Movimiento Celular , Proliferación Celular
7.
Cell Cycle ; 22(19): 2161-2171, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37946320

RESUMEN

LncRNAs are widely linked to the complex development of gastric cancer, which is acknowledged worldwide as the third highest contributor to cancer-related deaths and the fifth most common form of cancer. The primary focus of this study is to examine the role of LncRNA PSMG3-AS1 in a group of individuals with gastric cancer. The results of our study indicate that PSMG3-AS1 is highly expressed in over 20 different types of cancer. Significantly, there was a clear association found between the expression of PSMG3-AS1 and a multitude of TMB and MSI tumors. PSMG3-AS1 exhibited significant upregulation in gastric cancer patients compared to healthy individuals within the gastric cancer cohort. The prognosis of gastric cancer patients is intrinsically associated with PSMG3-AS1, as confirmed by survival analysis and ROC curves. Furthermore, we created a disruption vector based on LncRNA PSMG3-AS1 and introduced it into AGS and MKN-45 cells, which are human gastric cancer cells. Significant decreases in the expression of the PSMG3-AS1 gene were noticed in both intervention groups compared to the NC group, reflecting the protein level expressions. Significantly, the proliferative and invasive capabilities of MKN-45 and AGS cells were notably reduced following transfection with PSMG3-AS1 siRNA. The results of our study indicate that disruption of the LncRNA PSMG3-AS1 gene may impact the CAV1/miR-451a signaling pathway, thereby leading to a reduction in the ability of gastric cancer cells to multiply and invade.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , ARN Largo no Codificante/genética , MicroARNs/genética , ARN Interferente Pequeño , Transducción de Señal/genética , Regulación Neoplásica de la Expresión Génica/genética , Proliferación Celular/genética , Línea Celular Tumoral
8.
Appl Immunohistochem Mol Morphol ; 31(5): 324-330, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36952588

RESUMEN

This study aimed to explore the correlation of Golgi phosphoprotein 3 (GOLPH3) levels in papillary thyroid carcinoma (PTC) and papillary thyroid microcarcinoma (PTMC) with clinicopathologic features. GOLPH3 expression was determined by western blotting in solid tumors and the adjacent normal thyroid tissues. Mammalian target of rapamycin (mTOR) and Ki-67 were examined by immunohistochemical staining. Significantly higher levels of GOLPH3 protein were observed in PTC and PTMC compared with the adjacent normal thyroid tissues ( P <0.001). GOLPH3 level was positively associated with lymph node metastasis and clinical stage in PTC ( P <0.05) and utterly related to the clinical stage in PTMC ( P =0.012). No correlation was observed between GOLPH3 level and other clinicopathologic parameters such as sex, local invasion, tumor number, and tumor size. The expression level of GOLPH3 protein in mTOR-positive PTC was significantly higher than in mTOR-negative PTC ( P =0.002 in PTC, P =0.022 in PTMC) and positively correlated with Ki-67 proliferation index in PTC via Pearson correlation analysis ( r =0.353, P =0.007 in PTC; r =0.583, P <0.001 in PTMC). In conclusion, the relative expression level of GOLPH3 protein was significantly higher in PTC and PTMC than in normal thyroid tissues and increased with cancer severity. It may provide adjunctive information for diagnosing and predicting prognosis in patients with PTC or PTMC.


Asunto(s)
Relevancia Clínica , Neoplasias de la Tiroides , Humanos , Cáncer Papilar Tiroideo/patología , Antígeno Ki-67 , Neoplasias de la Tiroides/metabolismo , Serina-Treonina Quinasas TOR , Fosfoproteínas , Proteínas de la Membrana
9.
Ecotoxicol Environ Saf ; 241: 113778, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36068737

RESUMEN

Organophosphate flame retardants (OPFRs) are alternatives to brominated flame retardants (BFRs) and have recently gained wide acceptance in various materials. For the treatment and prevention of diseases, it is also important to clarify the relationship between OPFRs and tumors, despite the fact that OPFRs are less toxic than BFRs. This research used the TCGA and CTD databases for transcriptome profiling and identifying OPFRs-related genes. GO and KEGG analyses suggested that OPFRs may be closely related to colorectal cancer (CRC), and genes correlated with OPFRs were significantly and differently expressed between tumor and normal group. Further, OPFRs-related genes were associated with a good prognosis in CRC patients. The deeper research demonstrated that one of the OPFRs-triphenyl phosphate could significantly increased the viability and proliferation of CRC cell lines compared with the control group. In addition, Our research also found that melatonin at 50 µM could significantly impact CRC cell proliferation and migration ability induced by TPP.


Asunto(s)
Neoplasias Colorrectales , Retardadores de Llama , Línea Celular , Neoplasias Colorrectales/genética , Retardadores de Llama/metabolismo , Retardadores de Llama/toxicidad , Humanos , Organofosfatos/metabolismo , Organofosfatos/toxicidad , Compuestos Organofosforados/toxicidad
10.
J Oncol ; 2022: 8259135, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35756081

RESUMEN

Background: Long noncoding RNAs (lncRNAs) have a vital function in tumor onset and progress. For instance, long intergenic noncoding RNA 00641 (LINC00641) has been linked to cancer modulation. Nonetheless, the precise biological roles of LINC00641 in colorectal cancer (CRC) remain elusive. Methods: The expression levels of LINC00641 as well as the docking sites for LINC00641 and miR-450b-5p were analyzed using public data resources and web-based analytic tools. The putative downstream targets of miR-450b-5p were also predicted. Next, we evaluated the biological functions and the contents of LINC00641 in CRC both in vivo and in vitro. We next explored the influence of LINC00641 on the growth, migration, and infiltration of CRC cells via cell proliferation, migration, and invasion experiments. Besides, qRT-PCR, western blotting, flow cytometry, luciferase enzyme reporter assay, and in vivo tumorigenicity assays were conducted. Results: Our results confirmed that LINC00641 was markedly upmodulated in CRC tissues and CRC cell lines, and the upmodulation was linked to poor survival. Notably, the proliferative and migratory abilities of HCT-116 and SW480 cells were significantly inhibited by the knockdown of LINC00641 both in vitro and in vivo, illustrating that LINC00641 exerted a tumor-promotion role in CRC. Mechanistically, LINC00641 could competitively bind miR-450b-5p, thereby expunging its inhibitory effect on GOLPH3 expression. Moreover, miR-450-5p and GOLPH3 were able to reverse LINC00641-mediated cellular processes. Conclusions: Overall, the findings of this study suggest that LINC00641 promotes the proliferative and migratory abilities of CRC through sponging the miR-450b-5p/GOLPH3 axis.

11.
J Oncol ; 2022: 7007718, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35096064

RESUMEN

OBJECTIVE: The activity of NEK6 is enhanced in several cancer cells, including colon adenocarcinoma (COAD) cells. However, there are few reports on the microRNA (miRNA/miR) regulation of NEK6. In this study, we aimed to investigate the effects of miRNAs targeting NEK6 in COAD cells. METHODS: Public data and online analysis sites were used to analyze the expression levels of NEK6 and miR-323a-3p in COAD tissues as well as the relationship between NEK6 or miR-323a-3p levels and survival in patients with COAD and to predict miRNAs targeting NEK6. Real-time polymerase chain reaction and western blotting were performed to determine the levels of NEK6 and miR-323a-3p in COAD cells. The targeting of NEK6 by miR-323a-3p was verified using a dual-luciferase reporter assay. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, 5-ethynyl-2'-deoxyuridine assay, propidium iodide (PI) staining, annexin V-fluorescein isothiocyanate/PI staining, and transwell assay were employed to test the proliferation, apoptosis, migration ability, and invasiveness of COAD cells. RESULTS: In COAD cells, NEK6 was highly expressed, whereas miR-323a-3p was expressed at low levels and negatively regulated NEK6. Upregulating the level of miR-323a-3p impaired the proliferation, migration, and invasion of COAD cells and promoted apoptosis, whereas supplementing NEK6 alleviated the damage of the proliferation, migration, and invasion of COAD cells caused by miR-323a-3p and inhibited miR-323a-3p-induced apoptosis. These findings indicate that miR-323a-3p regulates the proliferation, migration, invasion, and apoptosis of COAD cells by targeting NEK6. CONCLUSION: miR-323a-3p downregulates NEK6 in COAD cells; this provides a novel basis for further understanding the occurrence and development of COAD.

12.
Dis Markers ; 2021: 8880282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33680216

RESUMEN

BACKGROUND: Early diagnosis is very important for the clinical treatment of gastric cancer (GC) and colorectal cancer (CRC). We aimed to detect Golgi phosphoprotein 3 (GOLPH3) and evaluate its diagnostic value. MATERIALS AND METHODS: Serum concentrations of GOLPH3 were detected by ELISA in 136 CRC patients, 102 GC patients, and 50 healthy controls at the Second Affiliated Hospital of Fujian Medical University from June 2016 to December 2019. Serum concentrations of CEA and CA19-9 were detected by ECLIA. RESULTS: Serum concentrations of GOLPH3, CEA, and CA19-9 were higher in GC and CRC patients than in healthy controls (P < 0.001). Serum GOLPH3 concentrations were increased in GC and CRC patients with tumors greater than 5 cm, poor differentiation, greater depth of tumor invasion, and increased lymphatic and distant metastases (P < 0.05). In the GC and CRC groups, the AUCs of GOLPH3 were higher than those of CEA and CA19-9 (P < 0.05), while the AUCs of the marker combination were higher than those of GOLPH3 (P < 0.05), and postoperative serum GOLPH3 levels were lower than preoperative levels (P < 0.001). Serum GOLPH3 concentrations in CRC patients correlated positively with CEA and CA19-9 concentrations (P < 0.05). CONCLUSION: Serum GOLPH3 concentrations in GC and CRC patients are related to TNM stage. GOLPH3 may represent a novel biomarker for the diagnosis of GC and CRC. The combination of serum GOLPH3, CEA, and CA19-9 concentrations can improve diagnostic efficiency for GC and CRC. GOLPH3 is expected to become an indicator for the early diagnosis and evaluation of surgical effects.


Asunto(s)
Neoplasias Colorrectales/sangre , Proteínas de la Membrana/sangre , Neoplasias Gástricas/sangre , Antígenos de Carbohidratos Asociados a Tumores/sangre , Antígeno Carcinoembrionario/sangre , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias Gástricas/patología
13.
Integr Cancer Ther ; 19: 1534735420972477, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33289438

RESUMEN

Protosappanin B (PSB) is a key active component of Lignum Sappan extract. Although the antiproliferative effects of Lignum Sappan extract have been demonstrated in various cancer cells, relatively little is known about the effects of PSB on tumor progression. The aim of this study was to explore the anti-tumor effects of PSB on human colon cancer cells by regulation of intracellular signaling pathways and Golgi phosphoprotein 3 (GOLPH3) expression in vitro and in vivo. Our results showed that PSB effectively inhibited the viability and migration of SW620 cells and induced apoptosis, but had poor effect on HCT116 cells. Furthermore, PSB significantly reduced the expression of p-AKT, p-p70S6K, ß-catenin, and p-ERK1/2 proteins in SW620 cells, and this effect was reversed by the corresponding signaling pathway agonists. Interestingly, PSB could also suppress GOLPH3 expression of SW620 cells in a concentration-dependent manner, but SW620 cells transfected with lentiviral vectors overexpressing GOLPH3 can effectively resist the cytotoxic activity of PSB in vitro. The xenograft experiment of SW620 cells with LV-GOLPH3 confirmed that PSB distinctly inhibited the tumor growth via suppressing GOLPH3 expression. Collectively, these findings clarified a new anti-cancer mechanism of PSB through inhibition of GOLPH3 expression and intracellular signaling pathways in colon cancer cells. PSB may be a potential new drug for colon cancer.


Asunto(s)
Neoplasias del Colon , Oxocinas , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Células HCT116 , Humanos , Proteínas de la Membrana , Transducción de Señal
14.
Cancer Manag Res ; 12: 12185-12195, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33273855

RESUMEN

PURPOSE: LACTB, regulated by a variety of microRNAs (miRNAs), is proven to be a tumor suppressor. However, there are few reports that LACTB in colon cancer cells is regulated by miRNA. Therefore, the aim of this study was to explore the miRNAs that regulate LACTB in colon cancer. PATIENTS AND METHODS: Data from TCGA were analyzed in starBase and GEPIA2, and Western blot and quantitative PCR (qPCR) were used to detect the expression of LACTB in colon cancer cell lines. MiRNAs targeting LACTB were predicted by MicroT-CDS, starBase, miRDB, mirDIP, and DIANA. The relationship between LACTB and miRNA was explored by dual-luciferase assay. MTT, propidium iodide (PI), Western blot, Annexin V-FITC/PI Kit, qPCR and transwell assay were used to detect the changes in cell proliferation, cell cycle, autophagy, apoptosis, epithelial-to-mesenchymal transition (EMT), cell migration, and invasiveness in colon cancer cells that overexpressed miR-1276 and/or LACTB. RESULTS: The results showed that the LACTB mRNA level was lower and the miR-1276 level was higher in colon cancer than in normal tissue. MiR-1276 inhibited the expression of LACTB. Furthermore, overexpression of miR-1276 in colon cancer cells increased proliferation, migration, invasiveness and EMT, and decreased autophagy and apoptosis. Supplementing LACTB suppressed these effects of miR-1276. CONCLUSION: In conclusion, miR-1276, which may be a potential therapy for colon cancer, inhibits cell growth and promotes apoptosis by targeting LACTB in colon cancer cells.

15.
Cancer Biother Radiopharm ; 35(10): 753-759, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32379491

RESUMEN

Background: FoxO3a is a well-studied tumor suppressor gene in the forkhead transcriptional factor O (FoxO) subfamily and its downregulation is correlated with the occurrence of gastric cancer (GC). GC tissues had microRNA (miR)-372 upregulation, which has targeted relationship with 3'-untranslated region (3'-UTR) of FoxO3a gene. This study investigated if miR-372 plays a role in modulating FoxO3a expression, and affecting GC cell proliferation, apoptosis, and cisplatin (DDP) resistance. Materials and Methods: Dual luciferase reporter gene assay assessed the targeted regulation between miR-372 and FoxO3a. DDP-resistant cell lines MGC803/DDP and MKN28/DDP were compared for gene expression against parental cells. Cell proliferation was measured by Cell Counting Kit-8 (CCK-8) assay. Cultured cells were transfected with miR-372 mimic or miR-negative control (NC) to measure FoxO3a mRNA and protein expression. Cell apoptosis and proliferation were tested by flow cytometry and 5-Ethynyl-2'-deoxyuridine (EdU) staining, respectively. Results: miR-372 had a targeted relationship with FoxO3a mRNA. MGC803/DDP and MKN28/DDP cells had significantly elevated miR-372 level than parental cells, while Foxo3a mRNA or protein levels were significantly decreased. CCK-8 assay revealed significantly lower inhibitory activity on cell proliferation in drug-resistant cells. Compared with miR-NC group, miR-273 inhibitor transfected DDP-resistant cells had significantly increased Foxo3a expression, enhanced cell apoptosis, reduced proliferation, and drug resistance. Conclusions: miR-372 upregulation is associated with DPP resistance of GC cells. Downregulation of miR-372 can inhibit proliferation, facilitate apoptosis, and suppress DDP resistance of drug-resistant GC cells.


Asunto(s)
Cisplatino/farmacología , Proteína Forkhead Box O3/metabolismo , MicroARNs/metabolismo , Neoplasias Gástricas/metabolismo , Antineoplásicos/farmacología , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Resistencia a Antineoplásicos , Citometría de Flujo , Proteína Forkhead Box O3/genética , Humanos , MicroARNs/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Transfección , Regulación hacia Arriba
16.
Artículo en Inglés | MEDLINE | ID: mdl-32454851

RESUMEN

OBJECTIVE: Tenacissoside H (TDH) is a Chinese medicine monomer extracted from Marsdenia tenacissima extract (MTE), which has been confirmed to have antitumor effects, but its mechanism is still unclear. The aim of this study was to investigate the effect and mechanism of TDH on human colon cancer LoVo cell proliferation and migration and explore the correlation of TDH treatment with the expression of GOLPH3 and cell signaling pathways in LoVo cells. METHODS: LoVo cells were treated with TDH at 0.1, 1, 10, and 100 µg/mL for 24, 48, and 72 h. The proliferation rate of LoVo cells was evaluated by MTT assay. Recombinant plasmid p-CMV-2-GOLPH3 was constructed, and p-CMV-2-GOLPH3 and p-CMV-2 empty plasmids were transfected into LoVo cells by lipofection. Western blotting was used to detect the transfection efficiency and the expression of p-p70S6K, p70S6K, ß-catenin, and GOLPH3. The apoptosis rate was analyzed with Annexin V-FITC/PI double-staining method, and cell migration assessed by transwell assay. RESULTS: TDH inhibited the proliferation of LoVo cells in a concentration-dependent manner. The IC50 of TDH treatment in LoVo cells at 24, 48, and 72 h was 40.24, 13.00, and 5.73 µg/mL, respectively. TDH treatment significantly induced apoptosis and suppressed the viability and migration of human colon cancer LoVo cells. The effect of TDH on induction of apoptosis and inhibition of migration in LoVo cells decreased significantly after activating the PI3K/AKT/mTOR and Wnt/ß-catenin signaling pathways with agonists. Additionally, the expression of GOLPH3 protein downregulated significantly in LoVo cells under TDH treatment. Overexpression of the GOLPH3 gene increased the expression of key proteins in PI3K/AKT/mTOR and Wnt/ß-catenin signaling pathways and blocked the antitumor activity of TDH. CONCLUSION: Collectively, the present results indicated that TDH can inhibit the proliferation vitality of colon cancer LoVo cells through downregulating GOLPH3 expression and activity of PI3K/AKT/mTOR and Wnt/ß-catenin signaling pathways.

17.
Int J Oncol ; 53(3): 1183-1192, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30015866

RESUMEN

Golgi phosphorylated protein (GOLPH)3 is overexpressed in colorectal cancer tissues and promotes the proliferation of colon cancer cells. A previous study by the authors demonstrated that GOLPH3 was associated with poor prognosis in colorectal cancer. However, the association between GOLPH3 gene overexpression and resistance to platinum-based drugs in colon cancer remains unknown. In the present study, the association between GOLPH3 overexpression and resistance of HT29 colon cancer cells to cisplatin and the mechanism underlying the development of chemoresistance were investigated. HT29 cells were divided into five groups. The expression of GOLPH3 mRNA was measured in the control and siRNA transfection groups. Reverse transcription-quantitative polymerase chain reaction analysis, cell proliferation, colony formation assay, tumor sphere formation and apoptosis (Annexin V) assays, western blotting and a nude mouse tumorigenicity assay were performed. HT29 cells were resistant to 10 µM cisplatin treatment, whereas the expression of GOLPH3, P-glycoprotein, phosphorylated extracellular signal-regulated kinase (pERK)1/2 and ß-catenin protein was significantly upregulated compared with the control group. With cisplatin treatment, silencing GOLPH3 gene expression downregulated the expression of these proteins, reduced cell proliferation and tumorigenicity, induced apoptosis and reversed the resistance of HT29 cells to cisplatin. In addition, the change in pERK1/2 and ß-catenin expression demonstrated that the mechanism of GOLPH3 overexpression involved in cisplatin resistance was associated with activation of the mitogen-activated protein kinase/ERK and Wnt/ß­catenin signaling pathways in HT29 cells. The tumorigenicity experiment in nude mice also demonstrated that silencing GOLPH3 expression increased the sensitivity of HT29 cells to cisplatin in vivo. Therefore, overexpression of GOLPH3 may be involved in the resistance of HT29 colon cancer cells to cisplatin chemotherapy by activating multiple cell signaling pathways.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cisplatino/uso terapéutico , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Femenino , Silenciador del Gen , Células HT29 , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Esferoides Celulares , Regulación hacia Arriba , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Med Rep ; 17(1): 542-548, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115442

RESUMEN

The novel proto­oncogene Golgi phosphoprotein (GOLPH)3 is overexpressed in a variety of tumor tissues and is associated with poor prognosis. The authors previously demonstrated that GOLPH3 gene is overexpressed in colorectal cancer tissues and promotes the proliferation of colonic cancer cells by activating the phosphatidylinositol­3­kinase/protein kinase B/the mammalian target of rapamycin and Wnt/ß­catenin signaling pathways. However, to the best of the authors' knowledge, if and how the GOLPH3 gene is involved in inducing resistance to colonic cancer chemotherapy has not been reported. In the present study, the association between the overexpression of the GOLPH3 gene and resistance of HT29 colonic cancer cells to 5­fluorouracil (5­FU) was investigated. Following confirmation of the effective silencing of the GOLPH3 gene, proliferation and apoptosis of colonic cancer cells were detected by MTT assay, colony formation assay and flow cytometry, and then the mechanism of GOLPH3­induced resistance to 5­FU chemotherapy in colonic cancer cells was investigated by western blotting. The results demonstrated that the expression of phosphorylated (p)­glycoprotein and GOLPH3 was increased in HT29 cells following treatment with 5­FU, which resulted in the development of drug resistance. Silencing GOLPH3 increased the sensitivity of HT29 cells to 5­FU, reduced their tumorigenicity and partly reversed their resistance to 5­FU. The expression of p­extracellular signal­regulated kinase (pERK)1/2 and ß­catenin was decreased, which indicated that its mechanism was associated with the activation of the mitogen­activated protein kinase/ERK and Wnt/ß­catenin signaling pathways. Therefore, GOLPH3 may be a potential, novel target for reversing chemotherapy resistance in colon cancer.


Asunto(s)
Resistencia a Antineoplásicos/genética , Fluorouracilo/farmacología , Expresión Génica , Proteínas de la Membrana/genética , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células HT29 , Humanos , Transfección
19.
J Cancer ; 7(8): 928-34, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27313783

RESUMEN

OBJECTIVE: Overexpression of GOLPH3 in colorectal cancer tissue may promote cell proliferation and activate the Wnt signaling pathway. We investigated the correlation between GOLPH3 gene expression and the Wnt signaling pathway to explore the mechanism of the overexpression of GOLPH3 gene which promotes proliferation in human colon cancer cells. METHODS: We measured expression of GOLPH3 mRNA in the human colon cancer cell lines HCT116, HT29, SW480 and SW620 by RT-PCR, and the cells with the highest expression were selected and divided into four groups: negative control, GOLPH3 siRNA transfection (siRNA-GOLPH3), Akt inhibitor (Tricinbine), and glycogen synthase kinase (GSK)-3ß inhibitor (TWS119). After human colon cancer cells were transfected with siRNA-GOLPH3, we used RT-PCR to investigate the silencing effect of GOLPH3 gene. We assessed the activity of the Wnt signaling pathway in all groups using the Topflash method. Proliferation and apoptosis of colon cancer SW620 cells were detected by MTT assay, colony formation assay and flow cytometry. Expression of Golgi phosphoprotein (GOLPH)3, ß-catenin, GSK-3ß and pS9-GSK-3ß in cancer cells was determined by Western blotting. RESULTS: SW620 cells expressed the highest level of GOLPH3 mRNA, and the silence effect was good after they were transfected with siRNA-GOLPH3. The relative luminescence units (RLU) values in the experimental groups were significantly lower than in the negative control group (P<0.001). There was no significant difference in the RLU values among the experimental groups (P> 0.05). The growth inhibition ratio and apoptosis rate of cancer cells in each experimental group were significantly higher than those in the control group, and the cell colony count in the experimental group was significantly lower than in the control group (P<0.05). In addition, the RLU value, proliferation and apoptosis rate of cancer cells did not differ significantly between each two experimental groups. Western blotting showed that, compared with the control group, expression of ß-catenin and pS9-GSK3 proteins were significantly decreased in the experimental group. Expression of GSK-3ß in the experimental group did not different from that of the control group. CONCLUSIONS: Overexpression of GOLPH3 gene activated the Wnt signaling pathway, as well as increasing expression of ß-catenin, promoting proliferation and inhibiting apoptosis in human colon cancer cells. The mechanism of action was that overexpression of GOLPH3 gene activated Akt, which may also further activate the Wnt signaling pathway via GSK-3ß, and promote proliferation in human colon cancer cells.

20.
World J Gastroenterol ; 21(48): 13473-9, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26730158

RESUMEN

AIM: To investigate the effect of Golgi phosphorylation protein 3 (GOLPH3) expression on cell apoptosis, angiogenesis and prognosis in colorectal cancer (CRC). METHODS: The expression of GOLPH3 in CRC tissues and normal colorectal mucosae was determined by immunohistochemistry in 62 patients. In addition, immunohistochemistry was also carried out to detect the expression of vascular endothelial growth factor (VEGF), CD34 and microvessel density (MVD). Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay was used to determine the apoptotic index (AI). The Kaplan-Meier method was used to analyze the relationship between GOLPH3 expression and survival in another 123 CRC cases. RESULTS: Compared with normal colorectal mucosae, a notably higher level of GOLPH3 protein expression was identified in CRC tissues (53.2% vs 24.2%, P < 0.05). Positive GOLPH3 expression was significantly associated with tumor invasion depth, TNM stage, and lymph node metastasis (P = 0.001; P = 0.020; P = 0.020; P < 0.05, respectively), but not with tumor length, tumor site, and age (P = 0.363; P = 0.819; P = 0.599; P > 0.05, respectively). VEGF expression and MVD in GOLPH3-positive CRC was significantly higher than in GOLPH3-negative CRC (VEGF: 69.7% vs 31.0%; MVD: 21.45 ± 9.39 vs 14.24 ± 8.97; P < 0.05). GOLPH3 expression was negatively correlated with AI in CRC as shown by Spearman correlation analysis (r = -0.320, P < 0.05). The 5-year survival rate in GOLPH3-negative CRC (69.4%) was significantly higher than in GOLPH3-positive CRC (48.6%) (log-rank test, P < 0.05). CONCLUSION: High expression of GOLPH3 is found in CRC tissues. GOLPH3 expression may be a novel prognostic marker for CRC patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Colorrectales/química , Proteínas de la Membrana/análisis , Anciano , Antígenos CD34/análisis , Apoptosis , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Microvasos/química , Microvasos/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Neovascularización Patológica , Factores de Tiempo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA