Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Pharmacol Res ; 188: 106654, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36640858

RESUMEN

The application of immune checkpoint inhibitors and FGFR protein tyrosine kinase inhibitors have made a tremendous breakthrough in bladder cancer therapy. However, inadequate drug responses and drug resistance interfere with successful treatment outcomes. For a new drug to enter the market, there is a long development cycle with high costs and low success rates. Repurposing previously Food and Drug Administration (FDA)-approved medications and using novel drug discovery strategies may be an optimal approach. Homoharringtonine (HHT) has been used for hematologic malignancies for over 40 years in China and was approved by the FDA approximately 10 years ago. Many studies have demonstrated that HHT effectively inhibits the development of several types of solid tumors, although the underlying mechanisms of action are unclear. In this study, we investigated the mechanisms underlying HHT activity against bladder cancer growth. We first compared HTT with the drugs currently used clinically for bladder cancer treatment. HHT showed stronger inhibitory activity than cisplatin, carboplatin, and doxorubicin. Our in vitro and in vivo data demonstrated that HHT inhibited proliferation, colony formation, migration, and cell adhesion of bladder cancer cells and induced apoptosis and cell cycle arrest in the nanomolar concentration range. Furthermore, we revealed that HHT treatment could downregulate the MAPK/Erk and PI3k/Akt signaling pathways by inactivating the integrin α5/ß1-FAK/Src axis. HHT-induced activity reduced cell-ECM interactions and cell migration, thus suppressing tumor metastasis progression. Altogether, HHT shows enormous potential as an anticancer agent and may be applied as a combination treatment strategy for bladder cancer.


Asunto(s)
Integrina alfa5 , Neoplasias de la Vejiga Urinaria , Humanos , Homoharringtonina/farmacología , Integrina alfa5/farmacología , Preparaciones Farmacéuticas , Fosfatidilinositol 3-Quinasas , Integrina alfa5beta1 , Línea Celular Tumoral , Apoptosis , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
2.
Cell Mol Life Sci ; 79(12): 614, 2022 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-36456730

RESUMEN

After the identification of specific epidermal growth factor receptor (EGFR)-activating mutations as one of the most common oncogenic driver mutations in non-small cell lung cancer (NSCLC), several EGFR-tyrosine kinase inhibitors (EGFR-TKIs) with different clinical efficacies have been approved by various health authorities in the last two decades in targeting NSCLC harboring specific EGFR-activating mutations. However, most patients whose tumor initially responded to the first-generation EGFR-TKI developed acquired resistance. In this study, we developed a novel combination strategy, "antiADAM17 antibody A9(B8) + EGFR-TKIs", to enhance the efficacy of EGFR-TKIs. The addition of A9(B8) was shown to restore the effectiveness of erlotinib and overcome acquired resistance. We found that when A9(B8) antibody was treated with erlotinib or gefitinib, the combination treatment synergistically increased apoptosis in an NSCLC cell line and inhibited tumor growth in vivo. Interestingly, the addition of A9(B8) could only reduce the survival of the erlotinib-resistant NSCLC cell line and inhibit the growth of erlotinib-resistant tumors in vivo but not gefitinib-resistant cells. Furthermore, we revealed that A9(B8) overcame erlotinib resistance through the FOXO3a/FOXM1 axis and arrested the cell cycle at the G1/S phase, resulting in the apoptosis of cancer cells. Hence, this study establishes a novel, promising strategy for overcoming acquired resistance to erlotinib through the FOXO3a/FOXM1 axis by arresting the cell cycle at the G1/S phase.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib , Neoplasias Pulmonares , Humanos , Anticuerpos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Resistencia a Medicamentos , Receptores ErbB/genética , Clorhidrato de Erlotinib/farmacología , Proteína Forkhead Box M1 , Gefitinib/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética
3.
Ecotoxicol Environ Saf ; 255: 114725, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-36924558

RESUMEN

Tributyltin chloride (TBTCL) is a widely used fungicide and heat stabilizer in compositions of PVC. TBTCL has been detected in human bodies and potentially causes harmful effects on humans' thyroid, cardiovascular and other organs. As one of the first examples of endocrine disruptors, the toxicity effects of TBTCL on the male reproduction system have aroused concerns. However, the potential cellular mechanisms are not fully explored. In the current study, by using Sertoli cells, a critical regulator of spermatogenesis as a cell model, we showed that with 200 nM exposure for 24 h, TBTCL causes apoptosis and cell cycle arrest. RNA sequencing analyses suggested that TBTCL probably activates endoplasmic reticulum (ER) stress, and disrupts autophagy. Biochemical analysis showed that TBTCL indeed induces ER stress and the dysregulation of autophagy. Interestingly, activation of ER stress and inhibition of autophagy is responsible for TBTCL-induced apoptosis and cell cycle arrest. Our results thus uncovered a novel insight into the cellular mechanisms for TBTCL-induced toxicology in Sertoli cells.


Asunto(s)
Células de Sertoli , Compuestos de Trialquiltina , Masculino , Humanos , Compuestos de Trialquiltina/toxicidad , Glándula Tiroides , Espermatogénesis , Apoptosis , Estrés del Retículo Endoplásmico , Autofagia
4.
Semin Cancer Biol ; 58: 80-89, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31054326

RESUMEN

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare diseases with a prevalence that has been increasing over the past three decades. The unsatisfactory outcome of these diseases has encouraged a great amount of research attention and discussion. Currently, to the best of our knowledge, studies on transcriptome screening and molecular mechanisms of the pathogenesis of GEP-NETs are limited. In this review, we have summarized the signaling network of GEP-NETs and the recent meta-analysis-related studies, and we have discussed the potential research direction for GEP-NETs, especially based on computational analysis.


Asunto(s)
Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Transducción de Señal/genética , Neoplasias Gástricas/genética , Animales , Biología Computacional/métodos , Estudios de Factibilidad , Humanos , Metaanálisis como Asunto
5.
Cancer Sci ; 111(8): 2803-2813, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32449268

RESUMEN

Death-associated protein kinase 1 (DAPK) is a calcium/calmodulin kinase that plays a vital role as a suppressor gene in various cancers. Yet its role and target gene independent of p53 is still unknown in hepatocellular carcinoma (HCC). In this study, we discovered that DAPK suppressed HCC cell migration and invasion instead of proliferation or colony formation. Using a proteomics approach, we identified DEAD-box helicase 20 (DDX20) as an important downstream target of DAPK in HCC cells and critical for DAPK-mediated inhibition of HCC cell migration and invasion. Using integrin inhibitor RGD and GTPase activity assays, we discovered that DDX20 suppressed HCC cell migration and invasion through the CDC42-integrin pathway, which was previously reported as an important downstream pathway of DAPK in cancer. Further research using cycloheximide found that DAPK attenuates the proteasomal degradation of DDX20 protein, which is dependent on the kinase activity of DAPK. Our results shed light on new functions and regulation for both DAPK and DDX20 in carcinogenesis and identifies new potential therapeutic targets for HCC.


Asunto(s)
Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Proteína 20 DEAD-Box/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Neoplasias Hepáticas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Células HEK293 , Humanos , Invasividad Neoplásica/patología , Regulación hacia Arriba
6.
J Cancer ; 15(5): 1378-1396, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38356704

RESUMEN

Purpose: Cervical cancer is a significant public health concern, particularly in developing countries. Despite available treatment strategies, the prognosis for patients with locally advanced cervical cancer and beyond remains poor. Therefore, an accurate prediction model that can reliably forecast prognosis is essential in clinical setting. Programmed cell death (PCD) mechanisms are diverse and play a critical role in tumor growth, survival, and metastasis, making PCD a potential reliable prognostic marker for cervical cancer. Methods: In this study, we created a novel prognostic indicator, programmed cell death-index (PCDi), based on a 10-fold cross-validation framework for comprehensive analysis of PCD-associated genes. Results: Our PCDi-based prognostic model outperformed previously published signature models, stratifying cervical cancer patients into two distinct groups with significant differences in overall survival prognosis, tumor immune features, and drug sensitivity. Higher PCDi scores were associated with poorer prognosis. The nomogram survival model integrated PCDi and clinical characteristics, demonstrating higher prognostic prediction performance. Furthermore, our study investigated the immune features of cervical cancer patients and found that those with high PCDi scores had lower infiltrating immune cells, lower potential of T cell dysfunction, and higher potential of T cell exclusion. Patients with high PCDi scores were resistant to classic chemotherapy regimens, including cisplatin, docetaxel, and paclitaxel, but showed sensitivity to the inhibitor SB505124 and Trametinib. Conclusion: Our findings suggest that PCD-related gene signature could serve as a useful biomarker to reliably predict prognosis and guide treatment decisions in cervical cancer.

7.
J Cancer ; 15(11): 3612-3624, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38817879

RESUMEN

Background: Cervical cancer is the fourth most common cancer among women worldwide. Cervical cancer usually develops from human papillomavirus (HPV) infection, which leads to cervical intraepithelial neoplasia (CIN1/2/3) and eventually invasive cervical cancer. Therefore, early-screening and detection of cervical lesions are crucial for preventing and treating cervical cancer. However, different regions have different levels of medical resources and availability of diagnostic methods. There is a need to compare the efficiency of different methods and combinations for detecting cervical lesions and provide recommendations for the optimal screening and detection strategies. Methods: The current clinical methods for screening and detection of cervical lesions mainly include TruScreen (TS), Thinprep cytologic test (TCT), HPV testing, and colposcopy, but their sensitivity and specificity vary and there is no standard protocol recommended. In this study, we retrospectively reviewed 2286 female samples that underwent cervical biopsy and compared the efficiency of different methods and combinations for detecting cervical lesions. Results: HPV screening showed the highest sensitivity for identifying women with CIN2+ cervical lesions compared with other single methods. Our results also showed the importance and necessary of the secondary diagnostic test like TCT and TS as a triage method before colposcopy examination and guided biopsy. Conclusions: Our study provides recommendations for the optimal screening and detection strategies for cervical lesions in different regions with different levels of development. As a non-invasive, easily operated, and portable device, TS is a promising tool to replace TCT for detecting cervical lesions in the health care center with insufficient medical resources.

8.
Environ Pollut ; 360: 124536, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39029862

RESUMEN

Tri (2-Ethylhexyl) phosphate (TEHP), widely used as a fire retardant and plasticizer, has been commonly found in the environment. Its potential health-related risks, especially reproductive toxicity, have aroused concern. However, the potential cellular mechanisms remain unexplored. In this study, we aimed to investigate the molecular mechanisms underlying TEHP-caused cell damage in Sertoli cells, which play a crucial role in supporting spermatogenesis. Our findings indicate that TEHP induces apoptosis in 15P-1 mouse Sertoli cells. Subsequently, we conducted RNA sequencing analyses, which suggested that ER stress, autophagy, and MAPK-related pathways may participate in TEHP-induced cytotoxicity. Furthermore, we demonstrated that TEHP triggers ER stress, activates p38 MAPK, and inhibits autophagy flux. Then, we showed that the inhibition of ER stress or p38 MAPK activation attenuates TEHP-induced apoptosis, while the inhibition of autophagy flux is responsible for TEHP-induced apoptosis. These results collectively reveal that TEHP induces ER stress, activates p38, and inhibits autophagy flux, ultimately leading to apoptosis in Sertoli cells. These shed light on the molecular mechanisms underlying TEHP-associated testicular toxicity.

9.
Cancer Lett ; 587: 216622, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38246224

RESUMEN

Triptolide, a natural bioactive compound derived from herbal medicine Tripterygium wilfordii, has multiple biological activities including anti-cancer effect, which is being tested in clinical trials for treating cancers. However, the exact mechanism by which Triptolide exerts its cytotoxic effects, particularly its specific protein targets, remains unclear. Here, we show that Triptolide effectively induces cytotoxicity in gastric cancer cells by increasing reactive oxygen species (ROS) levels. Further investigations reveal that ROS accumulation contributes to the induction of Endoplasmic Reticulum (ER) stress, and subsequently autophagy induction in response to Triptolide. Meanwhile, this autophagy is cytoprotective. Interestingly, through activity-based protein profiling (ABPP) approach, we identify peroxiredoxins-2 (PRDX2), a component of the key enzyme systems that act in the defense against oxidative stress and protect cells against hydroperoxides, as direct binding target of Triptolide. By covalently binding to PRDX2 to inhibit its antioxidant activity, Triptolide increases ROS levels. Moreover, overexpression of PRDX2 inhibits and knockdown of the expression of PRDX2 increases Triptolide-induced apoptosis. Collectively, these results indicate PRDX2 as a direct target of Triptolides for inducing apoptosis. Our results not only provide novel insight into the underlying mechanisms of Triptolide-induced cytotoxic effects, but also indicate PRDX2 as a promising potential therapeutic target for developing anti-gastric cancer agents.


Asunto(s)
Diterpenos , Fenantrenos , Neoplasias Gástricas , Humanos , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Peroxirredoxinas/genética , Diterpenos/farmacología , Fenantrenos/farmacología , Autofagia , Apoptosis , Compuestos Epoxi/farmacología
10.
J Hazard Mater ; 448: 130785, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36860030

RESUMEN

Tributyltin chloride (TBTCL), a commonly used antiseptic substance, is commonly found in the environment. Human exposure to TBTCL through the consumption of contaminated seafood, fish, or drinking water has aroused concern. It is well-characterized that TBTCL has multiple detrimental effects on the male reproductive system. However, the potential cellular mechanisms are not fully elucidated. Here, we characterized molecular mechanisms of TBTCL-induced cell injury in Leydig cells, a critical supporter for spermatogenesis. We showed that TBTCL induces apoptosis and cell cycle arrest in TM3 mouse Leydig cells. RNA sequencing analyses revealed that endoplasmic reticulum (ER) stress and autophagy were potentially involved in TBTCL-induced cytotoxicity. We further showed that TBTCL causes ER stress and inhibited autophagy flux. Notably, the inhibition of ER stress attenuates not only TBTCL-induces autophagy flux inhibition but also apoptosis and cell cycle arrest. Meanwhile, the activation of autophagy alleviates, and inhibition of autophagy exaggerates TBTCL-induced apoptosis and cell cycle arrest flux. These results suggest that TBTCL-induced ER stress and autophagy flux inhibition contributed to apoptosis and cell cycle arrest in Leydig cells, providing novel understanding into the mechanisms of TBTCL-induced testis toxicity.


Asunto(s)
Autofagia , Células Intersticiales del Testículo , Animales , Humanos , Ratones , Masculino , Testículo , Estrés del Retículo Endoplásmico
11.
Cell Death Discov ; 8(1): 296, 2022 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-35752613

RESUMEN

Gastric neuroendocrine carcinoma (GNEC) is a common type of neuroendocrine carcinoma (NEC) with a poor prognosis and limited therapeutic options. The underlying mechanisms of chemoresistance in patients with GNEC and those with NEC are largely unknown, and thus, reliable biomarkers and therapeutic targets that could improve treatment outcomes in patients with NECs are lacking. The aim of this study was to identify specific targets and investigate their roles in GNEC progression and treatment resistance. Differentially expressed genes (DEGs) were identified in GNEC specimens and were further analysed by focusing on their roles in chemoresistance. Gene Ontology (GO) and pathway enrichment analyses of GNEC DEGs revealed that synapse-related function was the most prominent cellular function perturbed in GNEC. SNAP25 was identified as the target gene involved in most of the enriched pathways. In vitro and in vivo experiments showed that SNAP25 plays a role in proliferation and chemoresistance in GNEC cell lines. AKT has been identified as a downstream target, and SNAP25 binds to AKT protein and promotes AKT protein half-life. Further analysis of other types of NEC as well as small cell lung cancer, which resembles NEC on a molecular level, has identified RUNDC3A as an upstream molecule that regulates SNAP25 expression and the associated phenotypes that could enhance chemoresistance in NECs. Our results show that SNAP25 expression in GNEC is mediated by RUNDC3A and promotes GNEC progression and chemoresistance via posttranslational modification of AKT. Thus, our results suggest that the RUNDC3A/SNAP25/Akt axis could be a potential therapeutic target in GNEC.

12.
Comput Struct Biotechnol J ; 18: 2610-2620, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33033581

RESUMEN

Gastric neuroendocrine carcinoma (GNEC) is rare cancer detected in the stomach. Previously, we demonstrated that the poorer prognosis of GNEC patients compared with gastric adenocarcinoma (GAC) patients was probably due to the lack of response to chemotherapy. Thus, it is crucial to study the specific GNEC gene expression pattern and investigate chemoresistance mechanism of GNEC. The transcriptome of GNEC patients was compared with that of GAC patients using RNA-seq. The KEGG analysis was employed to explore the specific differential expression gene function enrichment pattern. In addition, the transcriptomes of two GNEC cell lines, ECC10 and ECC12, were also compared with those of two GAC cell lines, MGC-803 and AGS, using RNA-seq. Comparing patient samples and cell lines transcriptome data, we try to uncover the potential targets and pathways which may affect the chemoresistance of GNEC. By combing all transcriptome data, we identified 22 key genes that were specifically up-regulated in GNEC. This panel of genes probably involves in the chemoresistance of GNEC. From our current experimental data, NeuroD1, one of the 22 genes, is associated with the prognosis of GNEC patients. Knockdown of NeuroD1 enhanced the sensitivity to irinotecan of GNEC cell lines. Our research sheds light in identifying a panel of novel therapeutic target specifically for GNEC clinical treatment which has not been reported before.

13.
Biomolecules ; 9(4)2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30999631

RESUMEN

Death Associated Protein Kinase 1 (DAPK1) is an important signaling kinase mediating the biological effect of multiple natural biomolecules such as IFN-γ, TNF-α, curcumin, etc. DAPK1 is degraded through both ubiquitin-proteasomal and lysosomal degradation pathways. To investigate the crosstalk between these two DAPK1 degradation pathways, we carried out a screen using a set of ubiquitin E2 siRNAs at the presence of Tuberous Sclerous 2 (TSC2) and identified that the small ubiquitin-like molecule (SUMO) pathway is able to regulate the protein levels of DAPK1. Inhibition of the SUMO pathway enhanced DAPK1 protein levels and the minimum domain of DAPK1 protein required for this regulation is the kinase domain, suggesting that the SUMO pathway regulates DAPK1 protein levels independent of TSC2. Suppression of the SUMO pathway did not enhance DAPK1 protein stability. In addition, mutation of the potential SUMO conjugation sites on DAPK1 kinase domain did not alter its protein stability or response to SUMO pathway inhibition. These data suggested that the SUMO pathway does not regulate DAPK1 protein degradation. The exact molecular mechanism underlying this regulation is yet to be discovered.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sitios de Unión , Proteínas Quinasas Asociadas a Muerte Celular/química , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Células HCT116 , Células HEK293 , Humanos , Unión Proteica , Proteolisis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
14.
Comb Chem High Throughput Screen ; 20(5): 395-402, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28025934

RESUMEN

BACKGROUND: Cancer is a complex disease and a huge threat to human health. The prognosis for some cancer types such as breast cancer has improved dramatically over the past 50 years due to rapid development of surgery, chemotherapy and radiotherapy technologies. However, for many patients bearing unresectable cancers, the prognosis remained poor. For this reason, there is always high demand for newer and better therapeutic reagents in cancer field. But the development of anticancer drugs is risky, lengthy and costly. Not many new anticancer reagents come into market every year. OBJECTIVE: This review focuses on the latest progress of chemotherapy drug repurposing in cancer research. CONCLUSION: It is clear from the review of that drug repurposing is faster and cheaper than conventional drug development process. The anticancer efficacy of numerous non-cancer drugs are being tested now. However, not many of them have obtained enough evidence to go into clinical trials for cancer treatment. How to filter the right candidate and narrow the gap between bench and bed remains a hurdle for both conventional and repurposing drug development.


Asunto(s)
Antineoplásicos/uso terapéutico , Reposicionamiento de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Humanos , Terapia Molecular Dirigida/métodos , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Mol Med Rep ; 15(2): 941-947, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28035421

RESUMEN

Sorafenib is a chemotherapeutic agent approved for the treatment of hepatocellular carcinoma (HCC) in China. Digitoxin is a cardiotonic drug, which has been demonstrated to exhibit anticancer effects in a number of cancers, but not in HCC. The aim of the present study was to evaluate the combinational effect of sorafenib and digitoxin on the treatment of HCC and to investigate the relevant molecular mechanisms of action that underlie these effects. The proliferation, cell death and migration of HCC cell lines, HepG2 and BEL­7402, were examined using MTT, acridine orange/ethidium bromide staining and scratch wound healing assays, respectively. In addition, alterations in the expression of phosphorylated-extracellular signal-regulated kinase (ERK), hypoxia­inducible factor 1­α (HIF­1α), hypoxia­inducible factor 2­α (HIF­2α) and vascular endothelial growth factor (VEGF) were measured prior to and following drug application using western blot analysis. Digitoxin and sorafenib synergistically inhibited cell viability, but did not inhibit migration, which was potentially mediated by suppression of ERK and hypoxia signaling. In downstream signaling pathways, the activity of ERK was synergistically suppressed by combinatorial treatment of HepG2 and BEL­7402 cells with sorafenib and digitoxin. In addition, the expression of HIF­1α, HIF­2α and VEGF was synergistically downregulated by combinational treatment.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Cardiotónicos/farmacología , Movimiento Celular/efectos de los fármacos , Digitoxina/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Hep G2 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Niacinamida/farmacología , Sorafenib , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
PLoS One ; 12(4): e0175290, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28388658

RESUMEN

Bladder cancer is one of the leading causes of cancer-related death in men, however, there was only limited effective treatment for invasive bladder cancer. DAPK1 has been shown to play important role in apoptosis and autophagy to suppress cancer progression. Previous results have shown that DAPK1 promoter was hypermethylated in the majority of bladder cancer specimens, however, the prognostic significance of DAPK1 in bladder cancer has yet to be demonstrated. In the present study, we found that DAPK1 expression was negatively associated with tumor stage and a low level expression of DAPK1 in bladder cancer specimens were associated with shorter survival in bladder cancer patients in 3 independent bladder cancer datasets (n = 462). Further investigation showed that FGFR3 knockdown resulted in downregulation of DAPK1 in bladder cancer cell line, suggesting that FGFR3 may be an upstream factor of DAPK1. Further analysis of the 3 independent bladder cancer datasets have identified ACOX1, UPK2, TRAK1, PLEKHG6 and MT1X genes had their expression significantly correlated with that of DAPK1. Knockdown of DAPK1 in bladder cancer T24 cells resulted in downregulation of ACOX1, UPK2 and TRAK1. Interestingly, TRAK1, by itself, was a favorable prognostic marker in the 3 independent bladder cancer datasets. Importantly, by using connectivity mapping with DAPK1-associated gene signature, we found that vemurafenib and trametinib could possibly reverse DAPK1-associated gene signature, suggesting that inhibition of Raf/MEK pathway may be a potential therapeutic approach for bladder cancer. Indeed, treatment of vemurafenib in T24 bladder cancer cells resulted in upregulation of DAPK1 confirming our connectivity mapping, while knockdown of DAPK1 resulted in reduced sensitivity towards inhibition of Braf signaling by vemurafenib. Together, our results suggest that DAPK1 is an important prognostic marker and therapeutic target for bladder cancer and have identified possible therapeutic agents for future testing in bladder cancer models with low DAPK1 expression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Neoplasias de la Vejiga Urinaria/enzimología
17.
World J Gastroenterol ; 23(11): 2012-2022, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28373767

RESUMEN

AIM: To evaluate the predictive value of the expression of chromosomal maintenance (CRM)1 and cyclin-dependent kinase (CDK)5 in gastric cancer (GC) patients after gastrectomy. METHODS: A total of 240 GC patients who received standard gastrectomy were enrolled in the study. The expression level of CRM1 and CDK5 was detected by immunohistochemistry. The correlations between CRM1 and CDK5 expression and clinicopathological factors were explored. Univariate and multivariate survival analyses were used to identify prognostic factors for GC. Receiver operating characteristic analysis was used to compare the accuracy of the prediction of clinical outcome by the parameters. RESULTS: The expression of CRM1 was significantly related to size of primary tumor (P = 0.005), Borrmann type (P = 0.006), degree of differentiation (P = 0.004), depth of invasion (P = 0.008), lymph node metastasis (P = 0.013), TNM stage (P = 0.002) and distant metastasis (P = 0.015). The expression of CDK5 was significantly related to sex (P = 0.048) and Lauren's classification (P = 0.011). Multivariate Cox regression analysis identified that CRM1 and CDK5 co-expression status was an independent prognostic factor for overall survival (OS) of patients with GC. Integration of CRM1 and CDK5 expression could provide additional prognostic value for OS compared with CRM1 or CDK5 expression alone (P = 0.001). CONCLUSION: CRM1 and CDK5 co-expression was an independent prognostic factors for GC. Combined CRM1 and CDK5 expression could provide a prognostic model for OS of GC.


Asunto(s)
Adenocarcinoma/mortalidad , Quinasa 5 Dependiente de la Ciclina/análisis , Carioferinas/análisis , Receptores Citoplasmáticos y Nucleares/análisis , Neoplasias Gástricas/mortalidad , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Femenino , Gastrectomía , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Carioferinas/metabolismo , Metástasis Linfática , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Receptores Citoplasmáticos y Nucleares/metabolismo , Estudios Retrospectivos , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Proteína Exportina 1
18.
J Cancer ; 7(9): 1049-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27326247

RESUMEN

Several previous studies have demonstrated that CDK5 or p27 expression in gastric cancer are associated with overall survival. We have previously reported that tumor suppressive function of CDK5 is related to p27. The aim of this study was to investigate correlation between the clinicopathological parameters and overall survival with different CDK5/p27 expression statuses in 244 gastric cancer patients using immunohistochemistry. Low CDK5 expression was detected in 93 cases (38.11%) and low p27 in 157 cases (64.34%). The expression of CDK5 was significantly related to sex (P = 0.034) and Lauren's classification (P = 0.013). The expression of p27 was significantly related to sex (P = 0.012), differentiation (P = 0.003), TNM stage (P = 0.013) and lymph node metastasis (P = 0.001). Based on the combined expression of CDK5 and p27, we classified the patients into four subtypes: CDK5 Low/p27 Low (n = 69), CDK5 High/p27 Low (n = 88), CDK5 Low/p27 High (n = 24) and CDK5 High/p27 High (n = 63). The CDK5 Low/p27 Low expression was closely related to female (P = 0.026), diffuse type (P = 0.027) and lymph node metastasis (P = 0.010). The CDK5 Low/p27 Low patients displayed poorer survival in comparison with the rest of the patients in Kaplan-Meier analysis. No significant overall survival difference was observed among the patients with CDK5 High and/or p27 High expression. In the multivariate analysis, CDK5 and p27 co-expression status was identified as an independent prognostic factor for patients with gastric cancer.

19.
J Cancer Res Clin Oncol ; 141(10): 1809-17, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25809905

RESUMEN

PURPOSE: Tumor stem cell surface marker CD44v6, a member of the CD44 protein family, is causally involved in the metastasis of cancer. Little is known about the functions of CD44v6 in gastric cancer. The aim of this study was to evaluate the prognostic value of CD44v6 and investigate its functional roles. METHODS: The expression of CD44v6 in 208 primary gastric adenocarcinoma patient samples was examined using immunohistochemistry and its correlation with clinicopathological parameters, and 5-year patient survival was assessed. Two pairs of MGC-803 stable cells with either CD44v6 overexpression or knockdown were created. The effect of CD44v6 on cell proliferation, colony formation, migration and apoptosis was investigated using these two pairs of cells. RESULTS: Overexpression of CD44v6 was observed in all cancer cell lines. The 5-year survival rate of patients with positive CD44v6 expression is significantly worse compared to those with negative expression (38.8 vs. 73.6 %). CD44v6 and TNM stage are two independent prognostic factors of primary gastric adenocarcinoma. The risk factors for the positive CD44v6 expression are location of tumor, depth of invasion, lymph node metastasis, Lauren classification and TNM stage. In MGC-803 cells, CD44 stimulated proliferation and colony formation, antagonized oxaliplatin-induced apoptosis, but did not affect migration. CONCLUSION: CD44v6 is an important prognosis marker in gastric cancer. Tissue specificity may affect the functions of CD44v6, and further work is needed to elucidate its regulation.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Apoptosis/fisiología , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Humanos , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pronóstico , Neoplasias Gástricas/mortalidad , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA