Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Prostate ; 82(4): 464-474, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35037281

RESUMEN

OBJECTIVES: This study sought to provide contemporary data from a multi-institution with respect to DNA-repair genes (DRGs) status and its impact on effects of platinum-based chemotherapy in treatment-emergent neuroendocrine prostate cancer (t-NEPC), for which little data exist. PATIENTS AND METHODS: All patients were retrospectively collected with eligible biopsied tissues for targeted next generation sequencing (NGS). The main outcomes were radiologic progression-free survival and overall survival according to Response Evaluation Criteria in Solid Tumors, version 1.1. RESULTS: Among the 43 NEPC patients, 13/43 (30%) harbored homozygous deletions, deleterious mutations, or both in DRGs. Eleven patients (11/13, 85%) with DRGs aberrations had effective response, including 7 patients with BRCA1/2 defects and 2 with mismatch repair-deficient caused by MSH2 alterations. While significantly fewer responders (30%) were detected in patients without DRGs aberrations (odds ratio = 12.83, p = 0.003). Compared with patients without genomic DRGs aberrations, the hazard ratio (HR) for radiologic progression in those with DRGs defects was 0.42 (95% confidence interval [CI]: 0.19-0.93), and the HR for death was 0.65 (95% CI: 0.24-1.72). The most common adverse event of Grade 3 or 4 was anemia, as noted in 7 patients (16%). CONCLUSION: The DRGs status is therapeutically meaningful in t-NEPC. Given the potential responses to platinum-based chemotherapy, our findings support the clinical use of NGS in t-NEPC patients to identify DRGs aberrations.


Asunto(s)
Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma Neuroendocrino/genética , Reparación del ADN/genética , Compuestos de Platino/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Anciano , Antineoplásicos , Proteína BRCA1/genética , Proteína BRCA2/genética , Carboplatino/uso terapéutico , Carcinoma Neuroendocrino/patología , Cisplatino/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/patología , Estudios Retrospectivos , Tasa de Supervivencia , Resultado del Tratamiento
2.
Nucleic Acids Res ; 47(8): 4211-4225, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30773595

RESUMEN

In PTEN-deficient prostate cancers, AKT signaling may be activated upon suppression of androgen receptor signaling. Activation of AKT as well as NF-κB signaling involves a key regulatory protein complex containing PHLPP, FKBP51 and IKKα. Here, we report a critical role of lncRNA PCAT1 in regulating the PHLPP/FKBP51/IKKα complex and progression of castration-resistant prostate cancer (CRPC). Using database queries, bioinformatic analyses, as well as RIP and RNA pull-down assays, we discovered and validated that the lncRNA-PCAT1 perturbs the PHLPP/FKBP51/IKKα complex and activates AKT and NF-κB signaling. Expression of lncRNA-PCAT1 is positively linked to CRPC progression. PCAT1 binds directly to FKBP51, displacing PHLPP from the PHLPP/FKBP51/IKKα complex, leading to activation of AKT and NF-κB signaling. Targeting PCAT1 restores PHLPP binding to FKBP1 leading to suppression of AKT signaling. Preclinical study in a mouse model of CRPC suggests therapeutic potential by targeting lncRNA PCAT1 to suppress CRPC progression. Together, the newly identified PCAT1/FKBP51/IKKα complex provides mechanistic insight in the interplay between AKT, NF-κB and AR signaling in CRPC, and the preclinical studies suggest that a novel role for PCAT1 as a therapeutic target.


Asunto(s)
Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , FN-kappa B/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Largo no Codificante/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Conjuntos de Datos como Asunto , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Masculino , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Br J Cancer ; 118(1): 88-97, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29123266

RESUMEN

BACKGROUND: Metastatic castration-resistant prostate cancer (mCRPC) is one of the main contributors to the death of prostate cancer patients. To date, the detailed molecular mechanisms underlying mCRPC are unclear. Given the crucial role of epithelial-mesenchymal transition (EMT) in cancer metastasis, we aimed to analyse the expression and function of Transforming growth factor-beta (TGF-ß) signal-associated protein named Sox5 in mCRPC. METHODS: The protein expression levels were analysed by western blot, immunohistochemistry and immunofluorescence. Luciferase reporter assays and chromatin immunoprecipitation were employed to validate the target of Sox5. The effect of Smad3/Sox5/Twist1 on PCa progression was investigated in vitro and in vivo. RESULTS: Here, we found that TGF-ß-induced EMT was accompanied by increased Sox5 expression. Interestingly, knockdown of Sox5 expression attenuated EMT induced by TGF-ß signalling. Furthermore, we demonstrated that Smad3 could bind to the promoter of Sox5 and regulate its expression. Mechanistically, Sox5 could bind to Twist1 promoter and active Twist1, which initiated EMT. Importantly, knockdown of Sox5 in prostate cancer cells resulted in less of the mesenchymal phenotype and cell migration ability. Furthermore, targeting Sox5 could inhibit prostate cancer progression in a xenograft mouse model. In clinic, patients with high Sox5 expression were more likely to suffer from metastases, and high Sox5 expression also has a lower progression-free survival and cancer specific-survival in clinic database. CONCLUSIONS: Therefore, we propose a new mechanism in which Smad3/Sox5/Twist1 promotes EMT and contributes to PCa progression.


Asunto(s)
Transición Epitelial-Mesenquimal , Proteínas Nucleares/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Factores de Transcripción SOXD/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína 1 Relacionada con Twist/genética , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Factores de Transcripción SOXD/genética , Transducción de Señal , Proteína smad3/metabolismo , Análisis de Supervivencia , Proteína 1 Relacionada con Twist/metabolismo , Regulación hacia Arriba
4.
Med Sci Monit ; 24: 8190-8197, 2018 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-30427826

RESUMEN

BACKGROUND Kid (kinesin-like DNA binding protein), a member of microtubule-dependent molecular motor proteins, also known as KIF22, is reported to be associated with carcinogenesis and cancer progression in different types of malignant tumor, but the biologic behavior and clinical outcome of KIF22 in prostate cancer (PCa) has not been well studied. This study aimed to analyze the association between KIF22 and clinical outcome in PCa patients. MATERIAL AND METHODS The expression of KIF22 in tumor specimens compared with paired paracancerous tissue from 114 patients undergoing radical prostatectomy was detected by immunohistochemistry; results were verified using The Cancer Genome Atlas (TCGA) database. Subsequently, the relationship between KIF22 expression and clinical prognosis of PCa patients was then statistically analyzed. RESULTS Both immunohistochemistry and database analysis showed that KIF22 was obviously overexpressed in PCa tissues compared with paracancerous tissue. The overexpression of KIF22 at the protein level was significantly related to higher clinical stage (P=0.025), Gleason score (P=0.002), seminal vesicle invasion (P=0.007), and lymph node metastasis (P=0.009). Furthermore, with the overexpression of KIF22 mRNA level in PCa patients, the oncological prognosis of PCa patients was much poorer. CONCLUSIONS High-level expression of KIF22 was related to both tumor progression and adverse clinical outcome. For this reason, KIF22 may become a potential prognostic factor for PCa.


Asunto(s)
Proteínas de Unión al ADN/sangre , Cinesinas/sangre , Neoplasias de la Próstata/metabolismo , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Cinesinas/genética , Cinesinas/metabolismo , Metástasis Linfática , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Pronóstico , Antígeno Prostático Específico/metabolismo , Prostatectomía , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Transcriptoma
5.
BMC Cancer ; 17(1): 530, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-28789622

RESUMEN

BACKGROUND: The independent prognostic role of squamous differentiation in pT1 bladder urothelial carcinoma has not been reported in previous studies. This article describes the impact of squamous differentiation on tumor recurrence and survival, and whether this histologic variant could indeed alter definitive treatment, based on single center-based retrospective data. METHODS: Totally, we retrieved (1)1449 histologically confirmed pT1 bladder urothelial carcinoma patients without histologic variants; (2)227 pT1 bladder urothelial carcinoma patients with squamous differentiation in our institution, from May 2004 to Oct 2015. The total amount of high/low grade urothelial carcinoma patients was 991/685 respectively. Transurethral resection of bladder tumor (TURBT) and intravesical chemotherapy were performed as initial treatments for all the patients. The clinical and pathological characteristics, treatment and survival outcomes were compared between squamous differentiation-positive and squamous differentiation-negative patients. RESULTS: In our study, 14% urothelial carcinoma patients were detected with squamous differentiation. The mean age of all the patients examined was 66.4, of whom 82% were males. The 5-year cancer specific survival rates were 69% for squamous differentiation-positive patients and 91% for squamous differentiation-negative patients (p < 0.001). Recurrence proved to be more common in squamous differentiation-positive patients than in negative patients. In the results of the univariate and multivariate Cox proportional hazard analysis, tumor size, lymphovascular invasion, recurrence and squamous differentiation were confirmed to be the prognostic factors associated with patients' survival. CONCLUSIONS: Squamous differentiation in pT1 bladder urothelial carcinoma is correlated to high risk of recurrence and poor prognosis as an independent prognostic factor. Radical cystectomy is essential for recurred high grade pT1 bladder urothelial carcinoma with squamous differentiation accompanied by lymphovascular invasion.


Asunto(s)
Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Carcinoma de Células Transicionales/mortalidad , Carcinoma de Células Transicionales/patología , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Transicionales/terapia , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Pronóstico , Modelos de Riesgos Proporcionales , Carga Tumoral , Neoplasias de la Vejiga Urinaria/terapia
6.
BMC Cancer ; 16: 180, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26940836

RESUMEN

BACKGROUND: Whether androgen deprivation therapy (ADT) leads to stroke morbidity is still unclear because of inconsistent evidence. We performed a systematic review and meta-analysis to evaluate if ADT used in men with prostate cancer (PCa) is associated with stroke. METHODS AND RESULTS: Medline, Embase and Cochrane Library databases up to September 30th 2014 were systematically searched with no date or language restriction, and reports from potentially relevant journals were complementally searched. Both randomized controlled trials and observational studies were included. Two reviewers independently extracted data and assessed study quality. Six observational studies finally met inclusion criteria, with 74,538 ADT users and 85,947 non-ADT users reporting stroke as an endpoint. Although no significant association was observed in pooled estimates, the incidence of stroke in ADT users was 12 % higher than control groups, (HR = 1.12, 95 % confidence interval [CI]: 0.95 to 1.32; P = 0.16). In subgroup-analyses of different ADT types, stroke was found to be significantly associated with gonadotropin-releasing hormone (GnRH) alone (HR = 1.20, 95 % CI: 1.12 to 1.28; P < 0.001), GnRH plus oral antiandrogen (AA) (HR = 1.23, 95 % CI: 1.13 to 1.34; P < 0.001) and orchiectomy (HR = 1.37, 95 % CI: 1.33 to 1. 46; P = 0.001), but not with AA alone (HR = 1.06, 95 % CI: 0.71 to 1.57; P = 0.78). CONCLUSIONS: GnRH alone, GnRH plus AA and orchiectomy is significantly associated with stroke in patients with PCa.


Asunto(s)
Antagonistas de Andrógenos/efectos adversos , Antineoplásicos Hormonales/efectos adversos , Hormona Liberadora de Gonadotropina/efectos adversos , Orquiectomía/efectos adversos , Neoplasias de la Próstata/complicaciones , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/etiología , Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Estudios de Casos y Controles , Hormona Liberadora de Gonadotropina/uso terapéutico , Humanos , Masculino , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/tratamiento farmacológico
7.
Tumour Biol ; 35(4): 3113-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24264313

RESUMEN

A HOXB13-to-IL17BR expression ratio was previously identified to predict a clinical outcome of breast cancer patients treated with adjuvant tamoxifen. A large number of studies were addressed to confirm its function as a predictor of breast cancer outcome treated with tamoxifen. However, conflicting results were got. In this study, a systematic search of databases was carried out, and other relevant papers were also identified. Then, the analyses were conducted according to the PRISMA and MOOSE guidelines. After full review, 11 studies with a total of 2,958 participants were deemed eligible and were included in the study. Pooled results revealed that women with higher HOXB13-to-IL17BR expression ratio had significantly worse outcomes in breast patients treated with tamoxifen, especially for those who are negative of node.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Homeodominio/genética , Receptores de Interleucina/genética , Tamoxifeno/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Femenino , Humanos , Receptores de Interleucina-17
8.
Tumour Biol ; 35(10): 9911-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24997620

RESUMEN

The aim of this study is to explore the oncological outcomes of hypofractionated radiotherapy for patients with prostate cancer. We systematically searched PubMed, Embase, and the Cochrane Library prior to April 2014 and references of relevant original papers and review articles. Unpublished data from meeting abstracts were supplemented. Studies comparing hypofractionated with conventionally fractionated radiotherapy (CFRT) on oncological outcomes of patients with clinically localized prostate cancer were included. Twelve distinct datasets involving 4,572 participants from 13 papers were eligible for this meta-analysis. The biochemical failure rate (BFR) decreases significantly in the hypofractionated radiotherapy (HFRT) group for approximately 30 % (11-43 %). However, no significantly lower prostate cancer-specific survival and overall survival in HFRT group were observed. In subgroup analyses, HFRT without radiation doses reduction was especially effective in controlling BFR (HR = 0.71, 95 % CI 0.53-0.95; P = 0.02). Compared to CFRT, HFRT only yield a consistent advantage on BFR in high-risk patients (HR = 0.61, 95 % CI 0.46-0.82; P = 0.001). Moreover, androgen deprivation therapy (ADT) had no effect on two radiotherapies in controlling BFR. HFRT improves biochemical failure-free survival in patients with clinically localized prostate cancer. Further well-designed trial is needed to confirm our findings.


Asunto(s)
Adenocarcinoma/radioterapia , Fraccionamiento de la Dosis de Radiación , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/radioterapia , Adenocarcinoma/mortalidad , Humanos , Masculino , Resultado del Tratamiento
9.
Sci Rep ; 14(1): 9411, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658579

RESUMEN

Matrix Metalloproteinases (MMPs) have been demonstrated to be essential in facilitating the migration and metastasis of clear cell renal cell carcinoma (ccRCC). However, the ability of the MMP family to predict clinical outcomes and guide optimal therapeutic strategies for ccRCC patients remains incompletely understood. In this investigation, we initially conducted a thorough examination of the MMP family in pan-cancer. Notably, MMPs exhibited distinctive significance in ccRCC. Following this, we undertook an extensive analysis to evaluate the clinical value of MMPs and potential mechanisms by which MMPs contribute to the progression of ccRCC. A novel stratification method and prognostic model were developed based on MMPs in order to enhance the accuracy of prognosis prediction for ccRCC patients and facilitate personalized treatment. By conducting multi-omics analysis and transcriptional regulation analysis, it was hypothesized that SAA1 plays a crucial role in promoting ccRCC migration through MMPs. Subsequently, in vitro experiments confirmed that SAA1 regulates ccRCC cell migration via the ERK-AP1-MMPs axis. In conclusion, our study has explored the potential value of the MMP family as prognostic markers for ccRCC and as guides for medication regimens. Additionally, we have identified SAA1 as a crucial factor in the migration of ccRCC.


Asunto(s)
Carcinoma de Células Renales , Movimiento Celular , Neoplasias Renales , Metaloproteinasas de la Matriz , Proteína Amiloide A Sérica , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/metabolismo , Humanos , Movimiento Celular/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Renales/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Pronóstico , Línea Celular Tumoral , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/genética , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Femenino , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Transducción de Señal
10.
Prostate ; 73(12): 1306-15, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23775364

RESUMEN

BACKGROUND: Cell-in-cell phenomenon has been found for more than a century. Entosis, which is a newly found homogeneous cell-in-cell phenomenon and a non-apoptosis cell death progress, has unclear function in prostate cancer progression. Here, we dissected mechanism of AR signaling related to entosis incidence in PCa progression. METHODS: Two stable PCa cell lines, named LNCaP-ARsi and C4-2-ARsi were established with stably transfected AR-shRNA to knockdown AR mRNA expression in LNCaP and C4-2 cells, respectively. PC3-AR9 cell line was also established after stably transfecting full-length AR-cDNA into PC3 cells. All these cells were cultured in poly-HEME-coated plates to induce entosis, which is demonstrated via double staining. RESULTS: Androgen-DHT could enhance entosis in LNCaP, C4-2 and PC3-AR9 PCa cells in a dose dependent manner. Knock-down of AR in LNCaP and C4-2 significantly suppressed entosis as compared to LNCaP-ARsc and C4-2-ARsc cells at both 1 and 10 nM DHT condition (P < 0.05). And suppression of Rho/ROCK expression resulted in interruption of AR-mediated entosis. Human PCa samples surveys demonstrated that entosis was found only in CRPC but not in BPH and ADPC where AR was less expressed as compared to CRPC. CONCLUSIONS: These results indicated that AR might play a negative role during PCa progression via influencing entosis by modulating Rho/ROCK pathway. This newly identified AR role of enhancing entosis might help us to better understand the multiple and opposite roles of AR, which could either promote or suppress PCa cell progression via different mechanisms.


Asunto(s)
Entosis/fisiología , Neoplasias de la Próstata/enzimología , Receptores Androgénicos/fisiología , Transducción de Señal/fisiología , Quinasas Asociadas a rho/fisiología , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/patología
11.
J Urol ; 189(3): 884-90, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23009871

RESUMEN

PURPOSE: Inguinal hernia is widely recognized as a complication after radical prostatectomy. We systematically investigated the risk factors for inguinal hernia, compared the incidence after various surgical procedures and explored prophylactic surgical maneuvers. MATERIALS AND METHODS: A systematic search of the literature was performed using Medline®, Web of Knowledge® and the Cochrane Library databases. All analyses and tests were conducted using STATA® software. RESULTS: A total of 31 trials from 29 eligible studies were identified according to the predefined selection criteria. As integrated, postoperative inguinal hernia developed in 15.9% (13.1-18.7) of patients who underwent radical retropubic prostatectomy and 6.7% (4.8-8.6) of those who underwent laparoscopic radical prostatectomy. Most cases of inguinal hernia occurred within the first 2 years after surgery. Right side and indirect-type dominance was found in those inguinal hernias. Pooled results of comparative studies revealed that the incidence of inguinal hernia after radical retropubic prostatectomy was significantly higher than that after no operation, laparoscopic surgery, radical perineal prostatectomy, mini-laparotomy radical retropubic prostatectomy and pelvic lymph node dissection, but was not significantly higher than that after open prostatectomy and cystectomy. In addition, increasing age, low body mass index, subclinical inguinal hernia, previous inguinal hernia repair and anastomotic stricture can increase the risk for inguinal hernia after radical prostatectomy. CONCLUSIONS: While some limitations cannot be overcome, this meta-analysis suggests that damage to the posterior layer of the rectus sheath may be involved in the development of inguinal hernia after radical prostatectomy. Prophylactic surgery for high risk subjects is advised at the time of radical prostatectomy to minimize the incidence of inguinal hernia.


Asunto(s)
Hernia Inguinal , Prostatectomía/efectos adversos , Prostatectomía/métodos , Neoplasias de la Próstata/cirugía , Salud Global , Hernia Inguinal/epidemiología , Hernia Inguinal/etiología , Hernia Inguinal/prevención & control , Humanos , Incidencia , Masculino , Factores de Riesgo
12.
BMC Cancer ; 13: 332, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23829273

RESUMEN

BACKGROUND: To explore the necessity of maintenance, efficacy of low-dose and superiority of various combination therapies of Bacillus Calmette-Guérin (BCG) in treatment of superficial bladder cancer (BCa). METHODS: Comprehensive searches of electronic databases (PubMed, Embase, and the Cochrane Library) were performed, then a systematic review and cumulative meta-analysis of 21 randomized controlled trials (RCTs) and 9 retrospective comparative studies were carried out according to predefined inclusion criteria. RESULTS: Significantly better recurrence-free survivals (RFS) were observed respectively in patients who received BCG maintenance, standard-dose and BCG plus epirubicin therapy comparing to those received induction, low-dose and BCG alone. BCG maintenance therapy was also associated with significantly better progression-free survival (PFS), but there were more incidences of adverse events. Pooled results showed no remarkable advantage of BCG combined with Mitomycin C or with interferon α-2b in improving oncologic outcomes. Sensitivity-analyses stratified by study-design and tumor stage led to very similar overall results and often to a decrease of the between-study heterogeneity. Our data confirmed that non-RCT only affected strength rather than direction of the overall results. CONCLUSIONS: All patients with superficial BCa should be encouraged to accept BCG maintenance therapy with standard-dose if well tolerated. Patients can benefit from BCG combined with epirubicin but not from BCG combined with Mitomycin C or interferon α-2b.


Asunto(s)
Antineoplásicos/uso terapéutico , Vacuna BCG/uso terapéutico , Carcinoma de Células Transicionales/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Administración Intravesical , Humanos
13.
Cancer Rep (Hoboken) ; 6(8): e1824, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37344930

RESUMEN

BACKGROUND: The six-transmembrane epithelial antigen of the prostate 3 (STEAP3) is a metalloreductase, which is essential for iron uptake. Existing literature has shown that STEAP3 may perform an important role in the onset and progression of tumors. Nonetheless, a complete pan-cancer investigation of the prognostic significance and immune properties of STEAP3 is currently unavailable. AIMS: As part of our investigation into the possible functions of STEAP3 in malignancies, we conducted a comprehensive analysis to examine the prognostic value and immune features of STEAP3 in human pan-cancer. METHODS AND RESULTS: R and Cytoscape programs were applied to analyze and visualize the data. The expression of STEAP3 in both cell lines and tissues was measured utilizing a variety of approaches. Using the shRNA knockdown technique, we tested the viability of the A498 and 786-O cell lines and validated their functions. Both CCK-8 and transwell assays were conducted to estimate cell proliferation and invasion. The expression of STEAP3 was substantially elevated and was shown to be linked to prognosis in the majority of malignancies, notably in clear cell renal cell carcinoma (ccRCC). In addition, the expression of STEAP3 was shown to have a strong correlation with immune infiltrates, which in turn triggered the recruitment and polarization of M2 macrophages in ccRCC. The protein STEAP3 shows promise as a predictor of responses to immune-checkpoint blockade (ICB) therapy. Positive links between STEAP3 and the epithelial-mesenchymal transition (EMT), the p53 pathway, and the immune-associated pathways were also found in the enrichment analysis. Our results illustrated that the STEAP3 expression level was substantially elevated in ccRCC tissues and suggested that it could stimulate EMT in ccRCC by downregulating CDH1. CONCLUSION: In a diverse range of cancers, STEAP3 might serve as a biomarker for determining the prognosis as well as a predictor of immunotherapy responsiveness. STEAP3 is a novel biological marker for determining prognosis, and it also performs a remarkable function in the promotion of tumor growth in ccRCC by enhancing invasion and EMT, as well as by triggering the recruitment and polarization of M2 macrophages.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Masculino , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Pronóstico , Próstata/patología , Proliferación Celular/genética
14.
J Exp Clin Cancer Res ; 42(1): 215, 2023 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-37599359

RESUMEN

BACKGROUND: N7-methylguanosine (m7G) modification is, a more common epigenetic modification in addition to m6A modification, mainly found in mRNA capsids, mRNA interiors, transfer RNA (tRNA), pri-miRNA, and ribosomal RNA (rRNA). It has been found that m7G modifications play an important role in mRNA transcription, tRNA stability, rRNA processing maturation, and miRNA biosynthesis. However, the role of m7G modifications within mRNA and its "writer" methyltransferase 1(METTL1) in tumors, particularly prostate cancer (PCa), has not been revealed. METHODS: The differential expression level of METTL1 between hormone-sensitive prostate cancer (HSPC) and castrate-resistant prostate cancer (CRPC) was evaluated via RNA-seq and in vitro experiments. The effects of METTL1 on CRPC progression were investigated through in vitro and in vivo assays. The upstream molecular mechanism of METTL1 expression upregulation and the downstream mechanism of its action were explored via Chromatin Immunoprecipitation quantitative reverse transcription polymerase chain reaction (CHIP-qPCR), Co-immunoprecipitation (Co-IP), luciferase reporter assay, transcriptome-sequencing, m7G AlkAniline-Seq, and mRNA degradation experiments, etc. RESULTS AND CONCLUSION: Here, we found that METTL1 was elevated in CRPC and that patients with METTL1 elevation tended to have a poor prognosis. Functionally, the knockdown of METTL1 in CRPC cells significantly limited cell proliferation and invasive capacity. Mechanistically, we unveiled that P300 can form a complex with SP1 and bind to the promoter region of the METTL1 gene via SP1, thereby mediating METTL1 transcriptional upregulation in CRPC. Subsequently, our findings indicated that METTL1 leads to enhanced mRNA stability of CDK14 by adding m7G modifications inside its mRNA, ultimately promoting CRPC progression.


Asunto(s)
Metiltransferasas , Neoplasias de la Próstata Resistentes a la Castración , Factor de Transcripción Sp1 , Humanos , Masculino , Proliferación Celular , Inmunoprecipitación de Cromatina , Quinasas Ciclina-Dependientes , Metiltransferasas/genética , MicroARNs , Neoplasias de la Próstata Resistentes a la Castración/genética , Estabilidad del ARN
15.
Cell Death Dis ; 14(3): 215, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36973255

RESUMEN

Enabled resistance or innate insensitiveness to antiandrogen are lethal for castration-resistant prostate cancer (CRPC). Unfortunately, there seems to be little can be done to overcome the antiandrogen resistance because of the largely unknown mechanisms. In prospective cohort study, we found that HOXB3 protein level was an independent risk factor of PSA progression and death in patients with metastatic CRPC. In vivo, upregulated HOXB3 contributed to CRPC xenografts progression and abiraterone resistance. To uncover the mechanism of HOXB3 driving tumor progression, we performed RNA-sequencing in HOXB3 negative (HOXB3-) and HOXB3 high (HOXB3 + ) staining CRPC tumors and determined that HOXB3 activation was associated with the expression of WNT3A and enriched WNT pathway genes. Furthermore, extra WNT3A and APC deficiency led HOXB3 to be isolated from destruction-complex, translocated to nuclei, and then transcriptionally regulated multiple WNT pathway genes. What's more, we also observed that the suppression of HOXB3 could reduce cell proliferation in APC-downregulated CRPC cells and sensitize APC-deficient CRPC xenografts to abiraterone again. Together, our data indicated that HOXB3 served as a downstream transcription factor of WNT pathway and defined a subgroup of CRPC resistant to antiandrogen which would benefit from HOXB3-targeted therapy.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Prospectivos , Genes Homeobox , Antagonistas de Andrógenos , Vía de Señalización Wnt , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
16.
Cell Death Dis ; 14(4): 289, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37095108

RESUMEN

As the most common modification of RNA, N6-methyladenosin (m6A) has been confirmed to be involved in the occurrence and development of various cancers. However, the relationship between m6A and castration resistance prostate cancer (CRPC), has not been fully studied. By m6A-sequencing of patient cancer tissues, we identified that the overall level of m6A in CRPC was up-regulated than castration sensitive prostate cancer (CSPC). Based on the analysis of m6A-sequencing data, we found m6A modification level of HRas proto-oncogene, GTPase (HRAS) and mitogen-activated protein kinase kinase 2 (MEK2 or MAP2K2) were enhanced in CRPC. Specifically, tissue microarray analysis and molecular biology experiments confirmed that METTL3, an m6A "writer" up-regulated after castration, activated the ERK pathway to contribute to malignant phenotype including ADT resistance, cell proliferation and invasion. We revealed that METTL3-mediated ERK phosphorylation by stabilizing the transcription of HRAS and positively regulating the translation of MEK2. In the Enzalutamide-resistant (Enz-R) C4-2 and LNCap cell line (C4-2R, LNCapR) established in the current study, the ERK pathway was confirmed to be regulated by METTL3. We also found that applying antisense oligonucleotides (ASOs) to target the METTL3/ERK axis can restore Enzalutamide resistance in vitro and in vivo. In conclusion, METTL3 activated the ERK pathway and induced the resistance to Enzalutamide by regulating the m6A level of critical gene transcription in the ERK pathway.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/patología , Andrógenos , Receptores Androgénicos/metabolismo , Resistencia a Antineoplásicos , Línea Celular Tumoral , Nitrilos , Proliferación Celular , Metiltransferasas
17.
Mol Biol Rep ; 39(4): 4429-37, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21938431

RESUMEN

The association between polymorphism of DNA methyltransferases 3B and cancer risk has been widely studied recently, and no consensus conclusion is available up to now. We perform a comprehensive search using the databases of Medline, ISI Web of Knowledge and Embase. The odds ratio (OR) and its 95% confidence interval (95% CI) are used to investigate the strength of the association. A total of 24 case-control studies with 15,647 individuals are included in this meta-analysis. For -149C > T (17 studies, 5229 cases and 6910 controls), no evidence indicate that individuals carrying the variant genotypes (CC + CT), relative to those carrying the wild homozygote TT genotype, have an increased risk of cancer (OR = 1.03; 95% CI = 0.84-1.26; P = 0.76). Similarly, no cancer risk is found in the subgroup analyses. For -579G > T (11 studies, 3513 cases and 3714 controls), significantly decreased risks of cancer are observed, and the ORs (95% CI) are 0.70 (0.56-0.87) for GT versus TT, 0.70 (0.57-0.85) for GG + GT versus TT and 0.76 (0.63-0.93) for G-allele versus T-allele, respectively. Subgroup analyses stratified by ethnicity and types of cancer are also performed, and results indicated that -579G > C polymorphism is associated with risk of cancer in Asians [0.68 (0.53-0.87) for GT vs. TT] but not in Europeans [0.82 (0.63-1.07) for GT vs. TT]. We also observe that the -579G is associated with decreased risk of colorectal cancer [0.49(0.38-0.62) for GT vs. TT]. More studies with larger sample size were needed to provide more precise evidence.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Predisposición Genética a la Enfermedad , Neoplasias/genética , Polimorfismo de Nucleótido Simple/genética , Pueblo Asiatico/genética , Estudios de Casos y Controles , Humanos , Oportunidad Relativa , Sesgo de Publicación , Factores de Riesgo , Población Blanca/genética , ADN Metiltransferasa 3B
18.
Urol Int ; 89(2): 233-40, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22890093

RESUMEN

OBJECTIVE: To examine the association between XPD and hOGG1 polymorphisms and prostate cancer (PCa) susceptibility. METHODS: A comprehensive search was conducted to identify all case-control studies on the relationship between XPD and hOGG1 polymorphisms and PCa risk. Odds ratios (ORs) were used to investigate the strength of the associations. RESULTS: A total of 15 case-control studies including 5,765 cases and 6,270 controls were eligible for the meta-analyses. For XPD Asp312Asn, no evidence indicated that individuals carrying 312Asn had an increased risk of PCa. In subgroup analyses, Asp312Asn polymorphism was associated with PCa risk in Asian populations [OR = 2.09 and 95% CI = 1.39-3.14 for Asn/Asn vs. Asp/Asp; OR = 1.49 and 95% CI = 1.12-1.98 for (Asn/Asn+Asn/Asp) vs. Asp/Asp]. For XPD Lys751Gln, the individuals with 751Gln did not have increased PCa risk compared with those with 751Arg, and no association was found in the subgroup analyses. For hOGG1 Ser326Cys, no significant association between the polymorphism and PCa risk was observed in the overall analysis. However, Ser326Cys was significantly associated with PCa risk under homologous contrast in Caucasians and Asians. CONCLUSIONS: XPD Asp312Asn polymorphism is associated with PCa risk in Asians and hOGG1 Ser326Cys polymorphism is associated with PCa risk in Caucasians and Asians.


Asunto(s)
ADN Glicosilasas/genética , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Proteína de la Xerodermia Pigmentosa del Grupo D/genética , Pueblo Asiatico , Estudios de Casos y Controles , Etnicidad , Frecuencia de los Genes , Genotipo , Homocigoto , Humanos , Masculino , Oportunidad Relativa , Polimorfismo Genético , Neoplasias de la Próstata/etnología , Riesgo , Población Blanca
19.
Oncogene ; 41(3): 387-399, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34759344

RESUMEN

Castration-resistant prostate cancer (CRPC) is a highly malignant type of advanced cancer resistant to androgen deprivation therapy. One of the important mechanisms for the development of CRPC is the persistent imbalanced regulation of AR and AR splice variants (AR/AR-Vs). In this study, we reported KDM4A-AS1, a recently discovered lncRNA, as a tumor promoter that was significantly increased in CRPC cell lines and cancer tissues. Depletion of KDM4A-AS1 significantly reduced cell viability, proliferation, migration in vitro, and tumor growth in vivo. We found that by binding to the NTD domain, KDM4A-AS1 enhances the stability of USP14-AR/AR-Vs complex, and promoted AR/AR-Vs deubiquitination to protect it from MDM2-mediated ubiquitin-proteasome degradation. Moreover, KDM4A-AS1 was found to enhance CRPC drug resistance to enzalutamide by repressing AR/AR-Vs degradation; antisense oligonucleotide drugs targeting KDM4A-AS1 significantly reduced the growth of tumors with enzalutamide resistance. Taken together, our results indicated that KDM4A-AS1 played an important role in the progression of CRPC and enzalutamide resistance by regulating AR/AR-Vs deubiquitination; targeting KDM4A-AS1 has broad clinical application potential.


Asunto(s)
Benzamidas/uso terapéutico , Histona Demetilasas con Dominio de Jumonji/metabolismo , Nitrilos/uso terapéutico , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Anciano , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Humanos , Masculino , Ratones , Nitrilos/farmacología , Feniltiohidantoína/farmacología
20.
Epigenomics ; 2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35852112

RESUMEN

Aims: We aimed to determine whether intronic circRNA acts as a molecular sponge in castration-resistant prostate cancer (CRPC). Materials & methods: A gene chip technique was used to conduct sequencing. A qPCR experiment was performed to verify the result. Radioimmunoprecipitation, RNA pull-down and dual-luciferase reporter assays were performed to particularly expound its function. Verification of downstream effects was carried out through qPCR and western blot, and a xenograft assay was performed in vivo for verification. Results: Intronic circRNA hsa_circ_0092339 in the nucleus was highly expressed in CRPC cell lines. hsa_circ_0092339 did not regulate the expression of its parental gene. hsa_circ_0092339 functions like a molecular sponge, preventing degradation of C-MYC mRNA by absorbing hsa-mir-940. Conclusion: hsa_circ_0092339 plays a critical role in CRPC through targeting C-MYC indirectly by absorbing hsa-mir-940.


Our research breaks the mold by investigating a novel function of RNA and a novel regulatory mechanism. Our research provides a new therapeutic target for prostate cancer treatment and broadens the understanding of prostate cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA